Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
0% found this document useful (0 votes)
249 views

Next Generation Sequencing Data Analysis

Uploaded by

Michael Guillien
Copyright
© © All Rights Reserved
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
249 views

Next Generation Sequencing Data Analysis

Uploaded by

Michael Guillien
Copyright
© © All Rights Reserved
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 435

i

Next-​Generation Sequencing
Data Analysis

Next-​generation DNA and RNA sequencing has revolutionized biology and


medicine. With sequencing costs continuously dropping and our ability to
generate large datasets rising, data analysis becomes more important than
ever. Next-​Generation Sequencing Data Analysis walks readers through next-​
generation sequencing (NGS) data analysis step by step for a wide range of
NGS applications.

For each NGS application, this book covers topics from experimental design,
sample processing, sequencing strategy formulation, to sequencing read quality
control, data preprocessing, read mapping or assembly, and more advanced
stages that are specific to each application. Major applications include:

• RNA-​seq: Both bulk and single cell (separate chapters)


• Genotyping and variant discovery through whole genome/​ exome
sequencing
• Clinical sequencing and detection of actionable variants
• De novo genome assembly
• ChIP-​seq to map protein-​DNA interactions
• Epigenomics through DNA methylation sequencing
• Metagenome sequencing for microbiome analysis

Before detailing the analytic steps for each of these applications, the book
presents introductory cellular and molecular biology as a refresher mostly
for data scientists, the ins and outs of widely used NGS platforms, and an
overview of computing needs for NGS data management and analysis. The
book concludes with a chapter on the changing landscape of NGS technolo-
gies and data analytics.

The second edition of this book builds on the well-​received first edition
by providing updates to each chapter. Two brand new chapters have been
added to meet rising data analysis demands on single-cell RNA-​seq and clin-
ical sequencing. The increasing use of long-reads sequencing has also been
reflected in all NGS applications. This book discusses concepts and principles
that underlie each analytic step, along with software tools for implementa-
tion. It highlights key features of the tools while omitting tedious details to
provide an easy-​to-​follow guide for practitioners in life sciences, bioinfor-
matics, biostatistics, and data science. Tools introduced in this book are open
source and freely available.
iii

Next-​Generation
Sequencing Data
Analysis
Second Edition

Xinkun Wang
iv

Second edition published 2024


by CRC Press
6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487-​2742
and by CRC Press
4 Park Square, Milton Park, Abingdon, Oxon, OX14 4RN
CRC Press is an imprint of Taylor & Francis Group, LLC
© 2024 Taylor & Francis Group, LLC
First edition published by CRC Press 2016
Reasonable efforts have been made to publish reliable data and information, but the author
and publisher cannot assume responsibility for the validity of all materials or the consequences
of their use. The authors and publishers have attempted to trace the copyright holders of all
material reproduced in this publication and apologize to copyright holders if permission to
publish in this form has not been obtained. If any copyright material has not been acknowledged
please write and let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted,
reproduced, transmitted, or utilized in any form by any electronic, mechanical, or other means,
now known or hereafter invented, including photocopying, microfilming, and recording, or in
any information storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, access www.
copyri​ght.com or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive,
Danvers, MA 01923, 978-​750-​8400. For works that are not available on CCC please contact
mpkbookspermissions@tandf.co.uk
Trademark notice: Product or corporate names may be trademarks or registered trademarks and
are used only for identification and explanation without intent to infringe.
ISBN: 9780367349899 (hbk)
ISBN: 9781032505701 (pbk)
ISBN: 9780429329180 (ebk)
DOI: 10.1201/​9780429329180
Typeset in Palatino
by Newgen Publishing UK
v

Contents

Preface to the Second Edition............................................................................... xv


Author....................................................................................................................xvii

Part I Introduction to Cellular and Molecular Biology

1. The Cellular System and the Code of Life................................................... 3


1.1 The Cellular Challenge............................................................................ 3
1.2 How Cells Meet the Challenge............................................................... 4
1.3 Molecules in Cells..................................................................................... 4
1.4 Intracellular Structures or Spaces........................................................... 5
1.4.1 Nucleus.......................................................................................... 5
1.4.2 Cell Membrane.............................................................................. 6
1.4.3 Cytoplasm...................................................................................... 7
1.4.4 Endosome, Lysosome, and Peroxisome.................................... 8
1.4.5 Ribosome....................................................................................... 8
1.4.6 Endoplasmic Reticulum.............................................................. 9
1.4.7 Golgi Apparatus........................................................................... 9
1.4.8 Cytoskeleton................................................................................ 10
1.4.9 Mitochondrion............................................................................ 10
1.4.10 Chloroplast.................................................................................. 12
1.5 The Cell as a System............................................................................... 12
1.5.1 The Cellular System................................................................... 12
1.5.2 Systems Biology of the Cell....................................................... 13
1.5.3 How to Study the Cellular System........................................... 14

2. DNA Sequence: The Genome Base............................................................. 17


2.1 The DNA Double Helix and Base Sequence....................................... 17
2.2 How DNA Molecules Replicate and Maintain Fidelity.................... 18
2.3 How the Genetic Information Stored in DNA Is Transferred
to Protein.................................................................................................. 20
2.4 The Genomic Landscape....................................................................... 21
2.4.1 The Minimal Genome................................................................ 21
2.4.2 Genome Sizes.............................................................................. 21
2.4.3 Protein-​Coding Regions of the Genome................................. 22
2.4.4 Non-​Coding Genomic Elements.............................................. 23
2.5 DNA Packaging, Sequence Access, and DNA-​Protein
Interactions.............................................................................................. 25
2.5.1 DNA Packaging.......................................................................... 25

v
vi Contents

2.5.2 Sequence Access......................................................................... 25


2.5.3 DNA-​Protein Interactions......................................................... 26
2.6 DNA Sequence Mutation and Polymorphism................................... 27
2.7 Genome Evolution.................................................................................. 28
2.8 Epigenome and DNA Methylation...................................................... 29
2.9 Genome Sequencing and Disease Risk................................................ 30
2.9.1 Mendelian (Single-​Gene) Diseases........................................... 31
2.9.2 Complex Diseases That Involve Multiple Genes................... 31
2.9.3 Diseases Caused by Genome Instability................................. 32
2.9.4 Epigenomic/​Epigenetic Diseases............................................. 32

3. RNA: The Transcribed Sequence................................................................. 35


3.1 RNA as the Messenger........................................................................... 35
3.2 The Molecular Structure of RNA.......................................................... 35
3.3 Generation, Processing, and Turnover of RNA as a
Messenger................................................................................................ 36
3.3.1 DNA Template............................................................................ 37
3.3.2 Transcription of Prokaryotic Genes......................................... 37
3.3.3 Pre-​mRNA Transcription of Eukaryotic Genes...................... 38
3.3.4 Maturation of mRNA................................................................. 40
3.3.5 Transport and Localization....................................................... 42
3.3.6 Stability and Decay.................................................................... 42
3.3.7 Major Steps of mRNA Transcript Level Regulation.............. 43
3.4 RNA Is More Than a Messenger.......................................................... 44
3.4.1 Ribozyme..................................................................................... 45
3.4.2 snRNA and snoRNA.................................................................. 46
3.4.3 RNA for Telomere Replication.................................................. 46
3.4.4 RNAi and Small Non-​Coding RNAs....................................... 47
3.4.4.1 miRNA......................................................................... 47
3.4.4.2 siRNA........................................................................... 49
3.4.4.3 piRNA.......................................................................... 49
3.4.5 Long Non-​Coding RNAs........................................................... 50
3.4.6 Other Non-​Coding RNAs......................................................... 50
3.5 The Cellular Transcriptional Landscape............................................. 51

Part II Introduction to Next-​Generation Sequencing


(NGS) and NGS Data Analysis

4. Next-​Generation Sequencing (NGS) Technologies: Ins and Outs........ 57


4.1 How to Sequence DNA: From First Generation to the Next............ 57
4.2 Ins and Outs of Different NGS Platforms........................................... 60
4.2.1 Illumina Reversible Terminator Short-​Read Sequencing..... 60
4.2.1.1 Sequencing Principle.................................................. 60
Contents vii

4.2.1.2 Implementation.......................................................... 60
4.2.1.3 Error Rate, Read Length, Data Output,
and Cost....................................................................... 63
4.2.1.4 Sequence Data Generation........................................ 63
4.2.2 Pacific Biosciences Single-​Molecule Real-​Time
(SMRT) Long-​Read Sequencing............................................... 64
4.2.2.1 Sequencing Principle.................................................. 64
4.2.2.2 Implementation.......................................................... 64
4.2.2.3 Error Rate, Read Length, Data Output,
and Cost....................................................................... 65
4.2.2.4 Sequence Data Generation........................................ 65
4.2.3 Oxford Nanopore Technologies (ONT) Long-​Read
Sequencing.................................................................................. 67
4.2.3.1 Sequencing Principle.................................................. 67
4.2.3.2 Implementation.......................................................... 68
4.2.3.3 Error Rate, Read Length, Data Output,
and Cost....................................................................... 68
4.2.3.4 Sequence Data Generation........................................ 69
4.2.4 Ion Torrent Semiconductor Sequencing.................................. 69
4.2.4.1 Sequencing Principle.................................................. 69
4.2.4.2 Implementation.......................................................... 70
4.2.4.3 Error Rate, Read Length, Date Output,
and Cost....................................................................... 70
4.2.4.4 Sequence Data Generation........................................ 72
4.3 A Typical NGS Workflow...................................................................... 72
4.4 Biases and Other Adverse Factors That May Affect NGS Data
Accuracy.................................................................................................. 74
4.4.1 Biases in Library Construction................................................. 74
4.4.2 Biases and Other Factors in Sequencing................................. 75
4.5 Major Applications of NGS................................................................... 76
4.5.1 Transcriptomic Profiling (Bulk and Single-​Cell
RNA-​Seq)..................................................................................... 76
4.5.2 Genetic Mutation and Variation Identification...................... 77
4.5.3 De Novo Genome Assembly...................................................... 77
4.5.4 Protein-​DNA Interaction Analysis (ChIP-​Seq)....................... 77
4.5.5 Epigenomics and DNA Methylation Study
(Methyl-​Seq)................................................................................ 77
4.5.6 Metagenomics............................................................................. 78

5. Early-​Stage Next-​Generation Sequencing (NGS) Data


Analysis: Common Steps............................................................................... 81
5.1 Basecalling, FASTQ File Format, and Base Quality Score................ 81
5.2 NGS Data Quality Control and Preprocessing................................... 84
5.3 Read Mapping......................................................................................... 86
5.3.1 Mapping Approaches and Algorithms.................................... 86
viii Contents

5.3.2 Selection of Mapping Algorithms and Reference


Genome Sequences..................................................................... 91
5.3.3 SAM/​BAM as the Standard Mapping File Format............... 93
5.3.4 Mapping File Examination and Operation............................. 95
5.4 Tertiary Analysis..................................................................................... 98

6. Computing Needs for Next-​Generation Sequencing (NGS)


Data Management and Analysis................................................................ 103
6.1 NGS Data Storage, Transfer, and Sharing......................................... 103
6.2 Computing Power Required for NGS Data Analysis...................... 105
6.3 Cloud Computing................................................................................ 106
6.4 Software Needs for NGS Data Analysis............................................ 108
6.4.1 Parallel Computing...................................................................110
6.5 Bioinformatics Skills Required for NGS Data Analysis...................111

Part III Application-​Specific NGS Data Analysis

7. Transcriptomics by Bulk RNA-​Seq............................................................117


7.1 Principle of RNA-​Seq............................................................................117
7.2 Experimental Design.............................................................................118
7.2.1 Factorial Design.........................................................................118
7.2.2 Replication and Randomization..............................................118
7.2.3 Sample Preparation and Sequencing Library
Preparation.................................................................................119
7.2.4 Sequencing Strategy................................................................. 121
7.3 RNA-​Seq Data Analysis....................................................................... 122
7.3.1 Read Mapping........................................................................... 122
7.3.2 Quantification of Reads........................................................... 126
7.3.3 Normalization........................................................................... 127
7.3.4 Batch Effect Removal............................................................... 129
7.3.5 Identification of Differentially Expressed Genes................. 129
7.3.6 Multiple Testing Correction.................................................... 133
7.3.7 Gene Clustering........................................................................ 134
7.3.8 Functional Analysis of Identified Genes............................... 134
7.3.9 Differential Splicing Analysis................................................. 136
7.4 Visualization of RNA-​Seq Data.......................................................... 137
7.5 RNA-​Seq as a Discovery Tool............................................................. 137

8. Transcriptomics by Single-Cell RNA-​Seq................................................ 145


8.1 Experimental Design............................................................................ 146
8.1.1 Single-Cell RNA-​Seq General Approaches........................... 146
8.1.2 Cell Number and Sequencing Depth..................................... 147
Contents ix

8.1.3 Batch Effects Minimization and Sample Replication.......... 149


8.2 Single-Cell Preparation, Library Construction, and
Sequencing............................................................................................ 150
8.2.1 Single-Cell Preparation............................................................ 150
8.2.2 Single Nuclei Preparation....................................................... 152
8.2.3 Library Construction and Sequencing.................................. 153
8.3 Preprocessing of scRNA-​Seq Data..................................................... 154
8.3.1 Initial Data Preprocessing and Quality Control................... 154
8.3.2 Alignment and Transcript Counting..................................... 156
8.3.3 Data Cleanup Post Alignment................................................ 157
8.3.4 Normalization........................................................................... 160
8.3.5 Batch Effects Correction........................................................... 162
8.3.6 Signal Imputation..................................................................... 164
8.4 Feature Selection, Dimension Reduction, and Visualization......... 165
8.4.1 Feature Selection....................................................................... 165
8.4.2 Dimension Reduction.............................................................. 166
8.4.3 Visualization.............................................................................. 168
8.5 Cell Clustering, Cell Identity Annotation, and Compositional
Analysis.................................................................................................. 171
8.5.1 Cell Clustering.......................................................................... 171
8.5.2 Cell Identity Annotation.......................................................... 173
8.5.3 Compositional Analysis........................................................... 177
8.6 Differential Expression Analysis........................................................ 178
8.7 Trajectory Inference.............................................................................. 181
8.8 Advanced Analyses.............................................................................. 185
8.8.1 SNV/​CNV Detection and Allele-​Specific Expression
Analysis...................................................................................... 185
8.8.2 Alternative Splicing Analysis................................................. 186
8.8.3 Gene Regulatory Network Inference..................................... 187

9. Small RNA Sequencing............................................................................... 205


9.1 Small RNA NGS Data Generation and Upstream
Processing.............................................................................................. 206
9.1.1 Data Generation........................................................................ 206
9.1.2 Preprocessing............................................................................ 207
9.1.3 Mapping..................................................................................... 208
9.1.4 Identification of Known and Putative Small
RNA Species.............................................................................. 209
9.1.5 Normalization........................................................................... 209
9.2 Identification of Differentially Expressed Small RNAs.................. 210
9.3 Functional Analysis of Identified Known Small RNAs.................. 210
x Contents

10. Genotyping and Variation Discovery by Whole Genome/​


Exome Sequencing........................................................................................ 215
10.1 Data Preprocessing, Mapping, Realignment, and
Recalibration......................................................................................... 216
10.2 Single Nucleotide Variant (SNV) and Short Indel Calling............. 217
10.2.1 Germline SNV and Indel Calling........................................... 217
10.2.2 Somatic Mutation Detection................................................... 219
10.2.3 Variant Calling from RNA Sequencing Data........................ 221
10.2.4 Variant Call Format (VCF)...................................................... 221
10.2.5 Evaluating VCF Results........................................................... 223
10.3 Structural Variant (SV) Calling........................................................... 225
10.3.1 Short-Read-​Based SV Calling................................................. 225
10.3.2 Long-Read-​Based SV Calling.................................................. 227
10.3.3 CNV Detection.......................................................................... 227
10.3.4 Integrated SV Analysis............................................................ 228
10.4 Annotation of Called Variants............................................................ 228

11. Clinical Sequencing and Detection of Actionable Variants................. 237


11.1 Clinical Sequencing Data Generation................................................ 238
11.1.1 Patient Sample Collection....................................................... 238
11.1.2 Library Preparation and Sequencing Approaches............... 240
11.2 Read Mapping and Variant Calling................................................... 243
11.3 Variant Filtering.................................................................................... 243
11.3.1 Frequency of Occurrence......................................................... 245
11.3.2 Functional Consequence......................................................... 245
11.3.3 Existing Evidence of Relationship to Human Disease........ 246
11.3.4 Clinical Phenotype Match....................................................... 246
11.3.5 Mode of Inheritance................................................................. 247
11.4 Variant Ranking and Prioritization.................................................... 247
11.5 Classification of Variants Based on Pathogenicity........................... 248
11.5.1 Classification of Germline Variants....................................... 248
11.5.2 Classification of Somatic Variants.......................................... 254
11.6 Clinical Review and Reporting.......................................................... 256
11.6.1 Use of Artificial Intelligence in Variant Reporting.............. 256
11.6.2 Expert Review........................................................................... 257
11.6.3 Generation of Testing Report.................................................. 257
11.6.4 Variant Validation..................................................................... 259
11.6.5 Incorporation into a Patient’s Electronic Health Record.... 260
11.6.6 Reporting of Secondary Findings.......................................... 260
11.6.7 Patient Counseling and Periodic Report Updates............... 260
11.7 Bioinformatics Pipeline Validation.................................................... 261

12. De Novo Genome Assembly with Long and/​or Short Reads............... 271
12.1 Genomic Factors and Sequencing Strategies for
De Novo Assembly................................................................................ 272
Contents xi

12.1.1 Genomic Factors That Affect De Novo Assembly................. 272


12.1.2 Sequencing Strategies for De Novo Assembly...................... 272
12.2 Assembly of Contigs............................................................................ 274
12.2.1 Sequence Data Preprocessing, Error Correction, and
Assessment of Genome Characteristics................................ 274
12.2.2 Contig Assembly Algorithms................................................. 277
12.2.3 Polishing.................................................................................... 280
12.3 Scaffolding and Gap Closure.............................................................. 281
12.4 Assembly Quality Evaluation............................................................. 282
12.5 Limitations and Future Development............................................... 284

13. Mapping Protein-​DNA Interactions with ChIP-​Seq............................. 293


13.1 Principle of ChIP-​Seq........................................................................... 293
13.2 Experimental Design............................................................................ 295
13.2.1 Experimental Control............................................................... 295
13.2.2 Library Preparation.................................................................. 295
13.2.3 Sequencing Length and Depth............................................... 296
13.2.4 Replication................................................................................. 296
13.3 Read Mapping, Normalization, and Peak Calling.......................... 297
13.3.1 Data Quality Control and Read Mapping............................ 297
13.3.2 Peak Calling.............................................................................. 300
13.3.3 Post-​Peak Calling Quality Control......................................... 307
13.3.4 Peak Visualization.................................................................... 309
13.4 Differential Binding Analysis............................................................. 310
13.5 Functional Analysis.............................................................................. 313
13.6 Motif Analysis....................................................................................... 314
13.7 Integrated ChIP-​Seq Data Analysis................................................... 315

14. Epigenomics by DNA Methylation Sequencing.................................... 321


14.1 DNA Methylation Sequencing Strategies......................................... 321
14.1.1 Bisulfite Conversion Methyl-​Seq............................................ 322
14.1.1.1 Whole-​Genome Bisulfite Sequencing
(WGBS)....................................................................... 322
14.1.1.2 Reduced Representation Bisulfite
Sequencing (RRBS)................................................... 323
14.1.2 Enzymatic Conversion Methyl-​Seq....................................... 324
14.1.3 Enrichment-​Based Methyl-​Seq............................................... 324
14.1.4 Differentiation of Cytosine Methylation from
Demethylation Products.......................................................... 325
14.2 DNA Methylation Sequencing Data Analysis.................................. 326
14.2.1 Quality Control and Preprocessing....................................... 326
14.2.2 Read Mapping........................................................................... 326
14.2.3 Quantification of DNA Methylation/​Demethylation
Products..................................................................................... 330
14.2.4 Visualization.............................................................................. 331
xii Contents

14.3 Detection of Differentially Methylated Cytosines


and Regions........................................................................................... 332
14.4 Data Verification, Validation, and Interpretation............................. 334

15. Whole Metagenome Sequencing for Microbial


Community Analysis.................................................................................... 341
15.1 Experimental Design and Sample Preparation................................ 342
15.1.1 Metagenome Sample Collection............................................. 343
15.1.2 Metagenome Sample Processing............................................ 343
15.2 Sequencing Approaches...................................................................... 344
15.3 Overview of Shotgun Metagenome Sequencing
Data Analysis........................................................................................ 345
15.4 Sequencing Data Quality Control and Preprocessing..................... 347
15.5 Taxonomic Characterization of a Microbial Community............... 347
15.5.1 Metagenome Assembly........................................................... 347
15.5.2 Sequence Binning..................................................................... 348
15.5.3 Calling of Genes and Other Genomic Elements from
Metagenomic Sequences......................................................... 351
15.5.4 Taxonomic Profiling................................................................. 351
15.6 Functional Characterization of a Microbial Community................ 352
15.6.1 Gene Function Annotation...................................................... 352
15.6.2 Gene Function Profiling and Metabolic Pathway
Reconstruction.......................................................................... 353
15.7 Comparative Metagenomic Analysis................................................ 354
15.7.1 Metagenome Sequencing Data Normalization.................... 354
15.7.2 Identification of Differentially Abundant
Species or OTUs........................................................................ 355
15.8 Integrated Metagenomics Data Analysis Pipelines......................... 355
15.9 Metagenomics Data Repositories....................................................... 355

Part IV The Changing Landscape of NGS


Technologies and Data Analysis

16. What’s Next for Next-​Generation Sequencing (NGS)?......................... 365


16.1 The Changing Landscape of Next-​Generation
Sequencing (NGS)................................................................................ 365
16.2 Newer Sequencing Technologies....................................................... 366
16.3 Continued Evolution and Growth of Bioinformatics
Tools for NGS Data Analysis.............................................................. 369
16.4 Efficient Management of NGS Analytic Workflows........................ 370
Contents xiii

16.5 Deepening Applications of NGS to Single-Cell and


Spatial Sequencing............................................................................... 372
16.6 Increasing Use of Machine Learning in NGS Data
Analytics................................................................................................ 374

Appendix I Common File Types Used in NGS Data Analysis.................. 383


Appendix II Glossary......................................................................................... 387
Index...................................................................................................................... 397
xv

Preface to the Second Edition

When I started working on the second edition of Next-​Generation Sequencing


Data Analysis, my primary goal was to add new chapters and contents on
clinical sequencing, single-cell sequencing, and third-​generation sequencing
(i.e., long reads) data analyses. These contents were either absent, or only
briefly discussed, in the first edition. For example, data processing for clinical
applications, where NGS has a direct impact on public health, was absent, and
a new chapter that covers clinical sequencing data QA/​QC, standard analysis
pipeline, and clinical interpretation is beneficial to the community. The dra-
matic growth in single-cell sequencing also warrants a new chapter, because
extracting rich biological information at the single-cell resolution requires
a new set of tools different from what is used to analyze “bulk” sequen-
cing data. Although long-read sequencing was covered in the first edition
of this book, technologies have since then made significant improvements
and achieved wide usage. Such developments require extensive updates to
nearly all of the applications, from RNA-​seq to metagenomics.
After meeting my primary goal, I set out to update the rest of the book.
This took much longer than I had initially planned. The challenges were two-​
fold. The first was updating the list of tools for each of the NGS applications.
Thanks to the productivity of the bioinformatics community, most NGS
applications have seen an abundance of tool development, and as a result
many new tools have emerged. Updating this large number of new tools took
quite some time. The second challenge was selectively introducing new and
existing tools, instead of overwhelming readers with a long list of tools that
have ever existed. While the tools presented in this edition are by no means
the most representative among all tools available, I made every effort to select
most of the effective open source tools in existence as of late 2022, drawing
information from benchmarking studies, citations, and recent updates.
In writing this edition, I have developed a renewed appreciation of the
intensity, excitement, and multiplicity of expertise in the NGS field. For
instance, there is an increasing convergence of expertise from artificial intel-
ligence, computer science, and high-​performance computing. At the same
time, because of the highly dynamic nature of the field, it becomes increas-
ingly challenging to keep abreast of the latest developments. This new
edition represents an effort from a practitioner in the field towards the goal of
informing readers on recent NGS data analysis tools. I would like to express
my gratitude to the many researchers and clinicians I have interacted with
in my role as the director of the Northwestern University NGS Facility. It is
their need for the latest NGS technologies that has kept me up to date with
the NGS field.

xv
newgenprepdf

xvi

Author

Xinkun Wang is Research Professor and the Director of the Next-​Generation


Sequencing Facility at Northwestern University in Chicago. Dr. Wang’s first
foray into the genomics field was during his doctoral training, performing
microarray-​based gene expression analysis. From 2005 to 2015, he was the
founding director of the University of Kansas Genomics Facility, prior to
moving to Northwestern to head the Northwestern University Sequencing
Facility (NUSeq) in late 2015. Dr. Wang is a renowned expert on genomics
technologies and data mining and their applications to the biomedical field.
Besides his monographic publications, he has published extensively in
neuroscience, with a focus on brain aging and neurodegenerative diseases
(mostly Alzheimer’s disease). Dr. Wang has served as principal investigator
on dozens of grants. Dr. Wang’s other professional activities include serving
on journal editorial boards, and as reviewers for journals, publishers, and
funding agencies.
Dr. Wang is a member of American Society of Human Genetics, Association
of Biomolecular Resource Facilities, the Honor Society of Phi Kappa Phi, and
Society for Neuroscience. Dr. Wang was born in Shandong province, China,
and is a first-​generation college graduate. His off-​work hobbies include cyc-
ling and Alpine skiing.

xvii
1

Part I

Introduction to Cellular and


Molecular Biology
1 
The Cellular System and the Code of Life

1.1 The Cellular Challenge


A cell, although minuscule with a diameter of less than 50 μm, works wonders if
you compare it to any human-​made system. Moreover, it perpetuates itself using
the information coded in its DNA. In case you ever had the thought of designing
an artificial system that shows this type of sophistication, you would know the
many insurmountable challenges such a system needs to overcome. A cell has
a complicated internal system, containing many types of molecules and parts.
To sustain the system, a cell needs to perform a wide variety of tasks, the most
fundamental of which are to maintain its internal order, prevent its system from
malfunctioning or breaking down, and reproduce or even improve the system,
in an environment that is constantly changing.
Energy is needed to maintain the internal order of the cellular system.
Without constant energy input, the entropy of the system will gradually
increase, as dictated by the second law of thermodynamics, and ultimately
lead to the destruction of the system. Besides energy, raw “building” material
is also constantly needed to renew its internal parts or build new ones if
needed, as the internal structure of a cell is dynamic and responds to constant
changes in environmental conditions. Therefore, to maintain the equilibrium
inside and with the environment, it requires a constant influx of energy and
raw material, and excretion of its waste. Guiding the capture of the requisite
energy and raw material for its survival and the perpetuation of the system is
the information encoded in its DNA sequence.
During the course of evolution a great number of organisms no longer
function as a single cell. The human body, for example, contains trillions
of cells. In a multicellular system, each cell becomes specialized to per-
form a specific function, e.g., β-​cells in our pancreas synthesize and release
insulin, and cortical neurons in the brain perform neurobiological functions
that underlie learning and memory. Despite this “division of labor,” the
challenges a single-​cell organism faces still hold true for each one of these
cells. Instead of dealing with the external environment directly, they interact
with and respond to changes in their microenvironment.

DOI: 10.1201/9780429329180-2 3
4 Next-Generation Sequencing Data Analysis

1.2 How Cells Meet the Challenge


Many cells, like algae and plant cells, directly capture energy from the sun or
other energy sources. Other cells (or organisms) obtain energy from the envir-
onment as heterotrophs. For raw material, cells can either fix carbon dioxide
in the air using the energy captured into simple organic compounds, which
are then converted to other requisite molecules, or directly obtain organic
molecules from the environment and convert them to requisite materials.
In the meantime, existing cellular components can also be broken down
when not needed for the re-​use of their building material. This process of
energy capture and utilization, and synthesis, interconversion, and breaking
down for re-​use of molecular material, constitutes the cellular metabolism.
Metabolism, the most fundamental characteristic of a cell, involves numerous
biochemical reactions.
Reception and transduction of various signals in the environment are crucial
for cellular survival. Reception of signals relies on specific receptors situated
on the cell surface, and for some signals, those inside the cell. Transduction
of incoming signals usually involves cascades of events in the cell, through
which the original signals are amplified and modulated. In response, cellular
metabolic profile is altered. The cellular signal reception and transduction
network is composed of circuits that are organized into various pathways.
Malfunctioning of these pathways can have a detrimental effect on cellular
response to the environment and eventually its survival.
Perpetuation and evolution of the cellular system rely on DNA replication
and cell division. The replication of DNA (to be detailed in Chapter 2) is a
high-​fidelity, but not error-​free, process. While maintaining the stability of
the system, this process also provides the mechanism for the diversification
and evolution of the cellular system. The cell division process is also tightly
regulated, for the most part to ensure equal transfer of the replicated DNA
into daughter cells. For the majority of multicellular organisms that repro-
duce sexually, in the process of germ cell formation the DNA is replicated
once but cell division occurs twice, leading to the reduction of DNA material
by half in the gametes. The recombination of DNA from female and male
gametes leads to further diversification in the offspring.

1.3 Molecules in Cells
Different types of molecules are needed to carry out the various cellular
processes. In a typical cell, water is the most abundant representing 70% of
the total cell weight. Besides water, there are a large variety of small and large
molecules. The major categories of small molecules include inorganic ions
The Cellular System and the Code of Life 5

(Na+​, K+​, Ca2+​, Cl-​, Mg2+​, etc.), monosaccharides, fatty acids, amino acids, and
nucleotides. Major varieties of large molecules are polysaccharides, lipids,
proteins, and nucleic acids (DNA and RNA). Among these components,
the inorganic ions are important for signaling (e.g., waves of Ca2+​ represent
important intracellular signal), cell energy storage (e.g., in the form of Na+​
/​K+​ cross-​membrane gradient), or protein structure/​function (e.g., Mg2+​ is
an essential cofactor for many metalloproteins). Carbohydrates (including
monosaccharides and polysaccharides), fatty acids, and lipids are major
energy-​providing molecules in the cell. Lipids are also the major component
of cell membrane. Proteins, which are assembled from 20 types of amino acids
in different order and length, underlie almost all cellular activities, including
metabolism, signal transduction, DNA replication, and cell division. They
are also the building blocks of many subcellular structures, such as cytoskel-
eton (see next section). Nucleic acids carry the code of life in their nearly
endless nucleotide permutations, which not only provides instructions on the
assembly of all proteins in cells but also exerts control on how such assembly
is carried out based on environmental conditions.

1.4 Intracellular Structures or Spaces


Cells maintain a well-​organized internal structure (Figure 1.1). Based on the
complexity of their internal structure, cells are divided into two major cat-
egories: prokaryotic and eukaryotic cells. The fundamental difference between
them is whether a nucleus is present. Prokaryotic cells, being more primor-
dial of the two, do not have a nucleus, and as a result their DNA is located in
a nucleus-​like but non-​enclosed area. Prokaryotic cells also lack organelles,
which are specialized and compartmentalized intracellular structures that
carry out different cellular functions (detailed next). Eukaryotic cells, on the
other hand, contain a distinct nucleus dedicated for DNA storage, mainten-
ance, and expression. Furthermore, they contain various organelles including
endoplasmic reticulum (ER), Golgi apparatus, cytoskeleton, mitochondrion,
and chloroplast (plant cells). The following is an introduction to the various
intracellular structures and spaces, including the nucleus, the organelles,
and other subcellular structures and spaces such as the cell membrane and
cytoplasm.

1.4.1 Nucleus
Since DNA stores the code of life, it must be protected and properly maintained
to avoid possible damage and ensure accuracy and stability. As proper execu-
tion of the genetic information embedded in the DNA is critical to the normal
functioning of a cell, gene expression must also be tightly regulated under
6 Next-Generation Sequencing Data Analysis

Nucleus
Nuclear Envelope
(with nuclear pores)
Cell Membrane
Chromatin
Peroxisome
Ribosome
Nucleolus Microtubule

Lysosome

Mitochondrion

Golgi Apparatus

Rough ER
Smooth ER

Intermediate
Filament

Centrosome
Cytoplasm Endosome
Microfilament

FIGURE 1.1
The general structure of a typical eukaryotic cell. Shown here is an animal cell.

all conditions. The nucleus, located in the center of most cells in eukaryotes,
offers a well-​protected environment for DNA storage, maintenance, and gene
expression. The nuclear space is enclosed by nuclear envelope consisting of
two concentric membranes. To allow movement of proteins and RNAs across
the nuclear envelope, which is essential for gene expression, there are pores
on the nuclear envelope that span the inner and outer membrane. The mech-
anical support of the nucleus is provided by the nucleoskeleton, a network
of structural proteins including lamins and actin among others. Inside the
nucleus, long strings of DNA molecules, through binding to certain proteins
called histones, are heavily packed to fit into the limited nuclear space. In
prokaryotic cells, a nucleus-​like irregularly shaped region that does not have
a membrane enclosure called the nucleoid provides a similar but not as well-​
protected space for DNA.

1.4.2 Cell Membrane
The cell membrane serves as a barrier to protect the internal structure of a
cell from the outside environment. Biochemically, the cell membrane, as well
as all other intracellular membranes such as the nuclear envelope, assumes
a lipid bilayer structure. While offering protection to their internal structure,
The Cellular System and the Code of Life 7

the cell membrane is also where cells exchange materials, and concurrently
energy, with the outside environment. Since the membrane is made of lipids,
most water-​soluble substances, including ions, carbohydrates, amino acids,
and nucleotides, cannot directly cross it. To overcome this barrier, there are
channels, transporters, and pumps, all of which are specialized proteins, on
the cell membrane. Channels and transporters facilitate passive movement,
that is, in the direction from high to low concentration, without consumption
of cellular energy. Pumps, on the other hand, provide active transportation of
the molecules, since they transport the molecules against the concentration
gradient and therefore consume energy.
The cell membrane is also where a cell receives most incoming signals from
the environment. After signal molecules bind to their specific receptors on the
cell membrane, the signal is relayed to the inside, usually eliciting a series of
intracellular reactions. The ultimate cellular response that the signal induces
is dependent on the nature of the signal, as well as the type and condition
of the cell. For example, upon detecting insulin in the blood via the insulin
receptor in their membrane, cells in the liver respond by taking up glucose
from the blood for storage.

1.4.3 Cytoplasm
Inside the cell membrane, cytoplasm is the thick solution that contains the
majority of cellular substances, including all organelles in eukaryotic cells
but excluding the nucleus in eukaryotic cells and the DNA in prokary-
otic cells. The general fluid component of the cytoplasm that excludes the
organelles is called the cytosol. The cytosol makes up more than half of the
cellular volume and is where many cellular activities take place, including
a large number of metabolic steps such as glycolysis and interconversion of
molecules, and most signal transduction steps. In prokaryotic cells, due to
the lack of the nucleus and other specialized organelles, the cytosol is almost
the entire intracellular space and where most cellular activities take place.
Besides water, the cytosol contains large amounts of small and large
molecules. Small molecules, such as inorganic ions, provide an overall bio-
chemical environment for cellular activities. In addition, ions such as Na+​,
K+​, and Ca2+​ also have substantial concentration differences between the
cytosol and the extracellular space. Cells spend a lot of energy maintaining
these concentration differences, and use them for signaling and metabolic
purposes. For example, the concentration of Ca2+​ in the cytosol is normally
kept very low at ~10−​7 M whereas in the extracellular space it is ~10−​3 M. The
rushing in of Ca2+​ under certain conditions through ligand-​or voltage-​
gated channels serves as an important messenger, inducing responses in a
number of signaling pathways, some of which lead to altered gene expres-
sion. Besides small molecules, the cytosol also contains large numbers of
macromolecules. Far from being simply randomly diffusing in the cytosol,
these large molecules form molecular machines that collectively function as
8 Next-Generation Sequencing Data Analysis

a “bustling metropolitan city” [1]. These supra-​macromolecular machines


are usually assembled out of multiple proteins, or proteins and RNA. Their
emergence and disappearance are dynamic and regulated by external and
internal conditions.

1.4.4 Endosome, Lysosome, and Peroxisome


Endocytosis is the process that cells bring in macromolecules, or other par-
ticulate substances such as bacteria or cell debris, into the cytoplasm from the
surroundings. Endosome and lysosome are two organelles that are involved
in this process. To initiate endocytosis, part of the cell membrane forms a
pit, engulfs the external substances, and then an endocytotic vesicle pinches
off from the cell membrane into the cytosol. Endosome, normally in the size
range of 300–​400 nm in diameter, forms from the fusion of these endocytotic
vesicles. The internalized materials contained in the endosome are sent to
other organelles such as lysosome for further digestion.
The lysosome is the principal site for intracellular digestion of internalized
materials as well as obsolete components inside the cell. Like the condition
in our stomach, the inside of the lysosome is acidic (pH at 4.5–​5.0), providing
an ideal condition for the many digestive enzymes within. These enzymes
can break down proteins, DNA, RNA, lipids, and carbohydrates. Normally
the lysosome membrane keeps these digestive enzymes from leaking into
the cytosol. Even in the event of these enzymes leaking out of the lysosome,
they can do little harm to the cell, since their digestive activities are heavily
dependent on the acidic environment inside the lysosome whereas the pH of
the cytosol is slightly alkaline (around 7.2).
Peroxisome is morphologically similar to the lysosome, but it contains
a different set of proteins, mostly oxidative enzymes that use molecular
oxygen to extract hydrogen from organic compounds to form hydrogen per-
oxide. The hydrogen peroxide can then be used to oxidize other substrates,
such as phenols or alcohols, via peroxidation reaction. As an example, liver
and kidney cells use these reactions to detoxify various toxic substances that
enter the body. Another function of the peroxisome is to break down long-​
chain fatty acids into smaller molecules by oxidation. Despite its important
functions, the origin of the peroxisome is still not entirely clear. One theory
proposes that this organelle has an endosymbiotic origin [2]. If this theory
holds true, all genes in the genome of the original endosymbiotic organism
must have been transferred to the nuclear genome. A more recent hypothesis,
however, is that they had an endogenous origin from the endomembrane
system, similar to the lysosome and the Golgi apparatus (see next section) [3].

1.4.5 Ribosome
Ribosome is the protein assembly factory in cells, translating genetic infor-
mation carried in messenger RNAs (mRNAs) into proteins. There are vast
The Cellular System and the Code of Life 9

numbers of ribosomes, usually from thousands to millions, in a typical cell.


While both prokaryotic and eukaryotic ribosomes are composed of two
components (or subunits), eukaryotic ribosomes are larger than their pro-
karyotic counterparts. In eukaryotic cells, the two ribosomal subunits are
first assembled inside the nucleus in a region called the nucleolus and then
shipped out to the cytoplasm. In the cytoplasm, ribosomes can be either
free, or get attached to another organelle (the ER). Biochemically, ribosomes
contain more than 50 proteins and several ribosomal RNA (rRNA) species.
Because ribosomes are highly abundant in cells, rRNAs are the most abun-
dant in total RNA extracts, accounting for 85% to 90% of all RNA species. For
profiling cellular RNA populations using next-​gen sequencing (NGS), rRNAs
are usually not of interest despite their abundance and therefore need to be
depleted to avoid generation of overwhelming amounts of sequencing reads
from them.

1.4.6 Endoplasmic Reticulum
As indicated by the name, ER is a network of membrane-​enclosed spaces
throughout the cytosol. These spaces interconnect and form a single internal
environment called the ER lumen. There are two types of ERs in cells: rough
ER and smooth ER. The rough ER is where all cell membrane proteins, such
as ion channels, transporters, pumps, and signal molecule receptors, as well
as secretory proteins, such as insulin, are produced and sorted. The charac-
teristic surface roughness of this type of ER comes from the ribosomes that
bind to them on the outside. Proteins destined for cell membrane or secre-
tion, once emerging from these ribosomes, are threaded into the ER lumen.
This ER-​targeting process is mediated by a signal sequence, or “address
tag,” located at the beginning part of these proteins. This signal sequence
is subsequently cleaved off inside ER before the protein synthesis process is
complete. Functionally different from the rough ER, the smooth ER plays an
important role in lipid synthesis for the replenishment of cellular membranes.
Besides membrane and secretory protein preparation and lipid synthesis,
one other important function of ER is to sequester Ca2+​ from the cytosol. In
Ca2+​-​mediated cell signaling, shortly after entry of the calcium wave into the
cytosol, most of the incoming Ca2+​ needs to be pumped out of the cell and/​or
sequestered into specific organelles such as ER and mitochondria.

1.4.7 Golgi Apparatus
Besides ER, the Golgi apparatus also plays an indispensable role in sorting
as well as dispatching proteins to the cell membrane, extracellular space,
or other subcellular destinations. Many proteins synthesized in the ER are
sent to the Golgi apparatus via small vesicles for further processing before
being sent to their final destinations. Therefore the Golgi apparatus is
10 Next-Generation Sequencing Data Analysis

sometimes metaphorically described as the “post office” of the cell. The pro-
cessing carried out in this organelle includes chemical modification of some
of the proteins, such as adding oligosaccharide side chains, which serves as
“address labels.” Other important functions of the Golgi apparatus include
synthesizing carbohydrates and extracellular matrix materials, such as the
polysaccharide for the building of the plant cell wall.

1.4.8 Cytoskeleton
Cellular processes like the trafficking of proteins in vesicles from ER to the
Golgi apparatus, or the movement of a mitochondrion from one intracellular
location to another, are not simply based on diffusion. Rather, they follow
certain protein-​made skeletal structure inside the cytosol, that is, the cyto-
skeleton, as tracks. Besides providing tracks for intracellular transport, the
cytoskeleton, like the skeleton in the human body, plays an equally important
role in maintaining cell shape, and protecting the cell framework from phys-
ical stresses as the lipid bilayer cell membrane is fragile and vulnerable to
such stresses. In eukaryotic cells, there are three major types of cytoskeletal
structures: microfilament, microtubule, and intermediate filament. Each type
is made of distinct proteins and has their own unique characteristics and
functions. For example, microfilament and microtubule are assembled from
actins and tubulins, respectively, and have different thickness (the diameter is
around 6 nm for microfilament and 23 nm for microtubule). While biochem-
ically and structurally different, both the microfilament and the microtubule
have been known to provide tracks for mRNA transport in the form of large
ribonucleoprotein complexes to specific intracellular sites, such as the distal
end of a neuronal dendrite, for targeted protein translation [4]. Besides its role
in intracellular transportation, the microtubule also plays a key role in cell
division through attaching to the duplicated chromosomes and moving them
equally into two daughter cells. In this process, all microtubules involved are
organized around a small organelle called a centrosome. Previously thought
to be only present in eukaryotic cells, cytoskeletal structure has also been
discovered in prokaryotic cells [5].

1.4.9 Mitochondrion
The mitochondrion is the “powerhouse” in eukaryotic cells. While some
energy is produced from the glycolytic pathway in the cytosol, most
energy is generated from the Krebs cycle and the oxidative phosphor-
ylation process that take place in the many mitochondria contained in a
cell. The number of mitochondria in a cell is ultimately dependent on its
energy demand. The more energy a cell needs, the more mitochondria
it has. Structurally, the mitochondrion is an organelle enclosed by two
membranes. The outer membrane is highly permeable to most cytosolic
The Cellular System and the Code of Life 11

molecules, and as a result the intermembrane space between the outer and
inner membranes is similar to the cytosol. Most of the energy releasing
process occurs in the inner membrane and in the matrix, that is, the space
enclosed by the inner membrane. For the energy release, high-​energy elec-
tron carriers generated from the Krebs cycle in the matrix are fed into an
electron transport chain embedded in the inner membrane. The energy
released from the transfer of high-​energy electrons through the chain to
molecular oxygen (O2), the final electron acceptor, creates a proton gra-
dient across the inner membrane. This proton gradient serves as the
energy source for the synthesis of ATP, the universal energy currency in
cells. In prokaryotic cells, since they do not have this organelle, ATP syn-
thesis takes place on their cytoplasmic membrane instead.
The origin of the mitochondrion, based on the widely accepted endo-
symbiotic theory, is an ancient α-Proteobacterium. So not surprisingly, the
mitochondrion carries its own DNA, but the genetic information contained
in the mitochondrial DNA (mtDNA) is extremely limited compared to the
nuclear DNA. The human mitochondrial DNA, for example, is 16,569 bp
in size coding for 37 genes, including 22 for transfer RNAs (tRNAs), 2
for rRNAs, and 13 for mitochondrial proteins. While it is much smaller
compared to the nuclear genome, there are multiple copies of mtDNA
molecules in each mitochondrion. Since cells usually contain hundreds
to thousands of mitochondria, there are a large number of mtDNA
molecules in each cell. In comparison, most cells only contain two copies
of the nuclear DNA. As a result, when sequencing cellular DNA samples,
sequences derived from mitochondrial DNA usually comprise a notable,
sometimes substantial, percentage of total generated reads. Although
small, the mitochondrial genomic system is fully functional and has the
entire set of protein factors for mtDNA transcription, translation, and
replication. As a result of its activity, when cellular RNA molecules are
sequenced, those transcribed from the mitochondrial genome also gen-
erate significant amounts of reads in the sequence output.
The many copies of mtDNA molecules in a cell may not all have the same
sequence due to mutations in individual molecules. Heteroplasmy occurs
when cells contain a heterogeneous set of mtDNA molecules. In general, mito-
chondrial DNA has a higher mutation rate than its nuclear counterpart. This
is because the transfer of high-​energy electrons along the electron transport
chain can produce reactive oxygen species as byproducts, which can oxidize
and cause mutations in mtDNA. To make this situation even worse, the DNA
repair capability in mitochondria is rather limited. Increased heteroplasmy
has been associated with higher risk of developing aging-​related diseases,
including Alzheimer’s disease, heart disease, and Parkinson’s disease [6].
Furthermore, mitochondrial DNA mutations have been known to underlie
aging and cancer development [7]. Certain hereditary mtDNA mutations
also underlie maternally inherited diseases that mostly affect the nervous
system and muscle, both of which are characterized by high energy demand.
12 Next-Generation Sequencing Data Analysis

1.4.10 Chloroplast
In animal cells, the mitochondrion is the only organelle that contains an
extranuclear genome. Plant and algae cells have another extranuclear genome
besides the mitochondrion, the plastid genome. Plastid is an organelle that can
differentiate into various forms, the most prominent of which is the chloroplast.
The chloroplast carries out photosynthesis through capturing the energy in sun-
light and fixing it into carbohydrates using carbon dioxide as substrate, and
releasing oxygen in the same process. For energy capturing, the green pigment
called chlorophyll first absorbs energy from sunlight, which is then transferred
through an electron transport chain to build up a proton gradient to drive the
synthesis of ATP. Despite the energy source, the buildup of proton gradient for
ATP synthesis in the chloroplast is very similar to that for ATP synthesis in the
mitochondrion. The chloroplast ATP derived from the captured light energy is
then spent on CO2 fixation. Similar to the mitochondrion, the chloroplast also
has two membranes: a highly permeable outer membrane and a much less per-
meable inner membrane. The photosynthetic electron transport chain, however,
is not located in the inner membrane, but in the membrane of a series of sac-​like
structures called thylakoids located in the chloroplast stroma (analogous to the
mitochondrial matrix).
Plastid is believed to be evolved from an endosymbiotic cyanobaterium,
which has gradually lost the majority of its genes in its genome over millions of
years. The current size of most plastid genomes is 120–​200 kb, coding for rRNAs,
tRNAs, and proteins. In higher plants there are around 100 genes coding for
various proteins of the photosynthetic system [8]. The transmission of plastid
DNA (ptDNA) from parent to offspring is more complicated than the maternal
transmission of mtDNA usually observed in animals. Based on the transmis-
sion pattern, it can be classified into three types: 1) maternal, inheritance only
through the female parent; 2) paternal, inheritance only through the male parent;
or 3) bioparental, inheritance through both parents [9]. Similar to the situation
in mitochondrion, there exist multiple copies of ptDNA in each plastid, and as
a result there are large numbers of ptDNA molecules in each cell with potential
heteroplasmy. Transcription from these ptDNA also generates copious amounts
of RNAs in the organelle. Therefore, sequence reads from ptDNA or RNA com-
prise part of the data when sequencing plant and algae DNA or RNA samples,
along with those from mtDNA or RNA.

1.5 The Cell as a System


1.5.1 The Cellular System
From the above description of a typical cell, it is obvious that the cell is a self-​
organizing system, containing many different molecules and structures that
The Cellular System and the Code of Life 13

work together coherently. Unlike other non-​biological systems, including


natural and artificial systems such as a car or a computer, the cell system is
unique as it continuously renews and perpetuates itself without violating
the laws of the physical world. It achieves this by obtaining energy from
and exchanging materials with its environment. The cellular system is also
characterized by its autonomy, that is, all of its activities are self-​regulated.
This autonomy is conferred by the genetic instructions coded in the cell’s
DNA. Besides such characteristics, the cell system is highly robust, as its
homeostasis is not easily disturbed by changes in its surroundings. This
robustness is a result of billions of years of evolution, which has led to the
building of tremendous complexity into the system. To study this complexity,
biologists have been mostly taking a reductionist approach to studying the
different cellular molecules and structures piece by piece. This approach has
been highly successful and much knowledge has been gathered on most parts
of the system. For a cell to function as a single entity, however, these different
parts do not work alone. To study how it operates as a whole, the different
parts need to be studied in the context of the entire system and therefore
a holistic approach is also needed. It has become more and more clear to
researchers in the life science community that the interactions between the
different cellular parts are equally, if not more, important as any part alone.

1.5.2 Systems Biology of the Cell


Systems biology is an emerging field that studies the complicated interactions
among the different parts of biological systems. It is an application of the
systems theory to the biological field. Introduced by the biologist Ludwig von
Bertalanffy in the 1940s, this theory aims to investigate the principles common
to all complex systems, and to describe these principles using mathematical
models. This theory is applicable to many disciplines including physics, soci-
ology, and biology, and one goal of this theory is to unify the principles of
systems as uncovered from the different disciplines. It is expected, therefore,
that principles uncovered from other systems may be applicable to biological
systems and provide guidance to better understanding of their working.
In the traditional reductionist approach, a single gene or protein is the
basic functioning unit. In systems biology, however, the basic unit is a
genetic circuit. Genetic circuit can be defined as a group of genes (or the
proteins they code) that work together to perform a certain task. There are
a multitude of tasks in a cell that need to be carried out by genetic circuits,
from the transduction of extracellular signal to the inside, the step-​by-​step
breakdown of energy molecules (such as glucose) to release energy, to the
replication of DNA prior to cell division. It is these genetic circuits that
underlie cellular behavior and physiology. If the information or material
flux in a genetic circuit is blocked or goes awry, the whole system will be
influenced, which might lead to the malfunctioning of the system and likely
a diseased state.
14 Next-Generation Sequencing Data Analysis

Based on the hierarchical organization principle of systems, gene circuits


interact with each other and form a complicated genetic network. Mapping
out a genetic network is a higher goal of systems biology. Genetic network
has been shown to share some common characteristics with non-​biological
networks such as the human society or the Internet [10]. One of such
characteristics is modularity, referring to the fact that genes (or proteins) that
work together to achieve a common goal often form a module and the module
is used as a single functional unit when needed. Another common character-
istic is the existence of hub or anchor nodes in the network, as a small number
of highly connected genes (or proteins) in a genetic network serve as hubs or
anchors through which other genes (or proteins) are connected to each other.

1.5.3 How to Study the Cellular System


Research into the systems biology of the cell is largely enabled by techno-
logical advancements in genomics, proteomics, and metabolomics. High-​
throughput genomics technologies, for example, allow simultaneous analysis
of tens of thousands of genes in an organism’s genome. Genome refers to the
whole set of genetic material in an organism’s DNA, including both protein-​
coding and non-​coding sequences. Similarly, proteome and metabolome are
defined as the complement of proteins and metabolites (small molecules),
respectively, in a cell or population of cells. Proteomics, through simultaneous
separation and identification of proteins in a proteome, provides answers to
the questions of how many proteins are present in the target cell(s) and at
what abundance levels. Metabolomics, on the other hand, through analyzing
a large number of metabolites simultaneously, monitors the metabolic status
of target cells.
The development of modern genomics technologies was mostly initiated
when the human genome was sequenced by the Human Genome Project.
The completion of the sequencing of this genome and the genomes of other
organisms, and the concurrent development of genomics technologies,
have for the first time offered an opportunity to study the systems prop-
erties of the cell. The first big wave of genomics technologies was mostly
centered on microarray, which enables analysis of the transcriptome and
subsequently study of genome-​ wide sequence polymorphism and the
epigenome. By studying all RNAs transcribed in a cell or population of
cells, transcriptomic analysis investigates what genes are active and how
active. Determination of genome-​wide sequence variations among indi-
viduals in a population enables examination of the relationship between
certain genomic polymorphisms and cellular dysfunctions, phenotypic
traits, or diseases. Epigenomic studies provide answers to the question
how the genomic information encoded in the DNA sequence is regulated
by the code conferred by chemical modifications of DNA bases. More
recently, the development of NGS technologies provides more power,
coverage, and resolution to the study of the genome, the transcriptome,
The Cellular System and the Code of Life 15

and the epigenome (for details on the development of NGS technologies


see Chapter 4). These NGS technologies, along with recent technological
developments in proteomics and metabolomics, further empower the
study of the cellular system.

References
1. Vale RD. The molecular motor toolbox for intracellular transport. Cell 2003,
112(4):467–​480.
2. de Duve C. Peroxisomes and related particles in historical perspective. Ann N
Y Acad Sci 1982, 386:1–​4.
3. Gabaldon T. Evolution of the peroxisomal proteome. Subcell Biochem 2018,
89:221–​233.
4. Das S, Vera M, Gandin V, Singer RH, Tutucci E. Intracellular mRNA transport
and localized translation. Nat Rev Mol Cell Biol 2021, 22(7):483–​504.
5. Mayer F. Cytoskeletons in prokaryotes. Cell Biol Int 2003, 27(5):429–​438.
6. Chocron ES, Munkacsy E, Pickering AM. Cause or casualty: the role of mito-
chondrial DNA in aging and age-​associated disease. Biochim Biophys Acta Mol
Basis Dis 2019, 1865(2):285–​297.
7. Smith ALM, Whitehall JC, Greaves LC. Mitochondrial DNA mutations in
ageing and cancer. Mol Oncol 2022, 16(18):3276–​3294.
8. de Vries J, Archibald JM. Plastid genomes. Curr Biol 2018, 28(8):R336–​R337.
9. Harris SA, Ingram R. Chloroplast DNA and biosystematics: the effects of
intraspecific diversity and plastid transmission. Taxon 1991:393–​412.
10. Roy U, Grewal RK, Roy S. Complex Networks and Systems Biology. In:
Systems and Synthetic Biology. Springer; 2015: 129–​150.
2 
DNA Sequence: The Genome Base

2.1 The DNA Double Helix and Base Sequence


Among the different types of molecules in cells, DNA has a structure that
makes it ideal to code the blueprint of life. The building blocks of DNA are
nucleotides, which are made up of three chemical groups: a five-​carbon sugar
(deoxyribose), phosphate, and one of four nucleobases. The spatial struc-
ture of DNA is a double helix comprised of two strands. The backbone of
each strand is made of the sugar moiety and phosphate, which are invari-
ably connected in an alternating fashion and therefore do not carry genetic
information. The “rungs” that connect the two strands are composed of
nucleobases, which are where the information is stored. Since the discovery
of this structure in 1953 by Watson and Crick, the elegance and simplicity of
this structure has fascinated generations of biologists, chemists, and scientists
from other fields.
There are four different types of nucleobases (or simply bases) in
DNA: two purines (adenine, usually abbreviated as A; and guanine, G),
and two pyrimidines (cytosine, C; and thymine, T). Nucleobases in the two
DNA strands that form the rung structure interact via hydrogen bonding in
a fixed manner: A always pairing with T, and C with G. This complementary
base-​pairing pattern enables the DNA molecule to assume the most thermo-
dynamically favorable structure. The fixed pairing pattern between the bases
makes it easy to provide coding for life and to replicate for perpetuation.
The almost endless arrangements of the base pairs in DNA provide the
basis for its role as the genetic information carrier. The information embedded
in the DNA base sequence dictates what, when, and how many proteins are
made in a cell at a certain point of time. At a deeper level, the information
codes for the entire operating logic of the cellular system. It contains all
instructions needed to form a new life and for it to grow, develop, and repro-
duce. From the medical point of view, alterations or polymorphisms in the
DNA base sequence can predispose us to certain diseases as well as underlie
our responses to medications.

DOI: 10.1201/9780429329180-3 17
18 Next-Generation Sequencing Data Analysis

2.2 How DNA Molecules Replicate and Maintain Fidelity


The DNA’s double helix structure and complementary base-​pairing make it
robust to copy the bio-​information it carries through its replication. To rep-
licate, the two strands of the parent DNA molecule are first unwound by an
enzyme called helicase. The two unwound strands then serve as templates
for the synthesis of new complementary strands, giving rise to two offspring
DNA molecules. The enzyme that carries out the new strand synthesis is called
DNA polymerase, which assembles nucleotides into a new strand by adding
one nucleotide at a time to a pre-​existing primer sequence based on comple-
mentary base-​pairing with the template strand (Figure 2.1). Biochemically, the
enzyme catalyzes the formation of a covalent phosphodiester bond between
the 5’-​phosphate group of the incoming complementary nucleotide and the
3’-​hydroxyl group on the elongating strand end. Besides elongating the new
DNA strand, most DNA polymerases also have proofreading capability. If
a nucleotide that is not complementary to the template is attached to the
end of the elongating strand (i.e., mis-​pairing), the enzyme will turn around
and cleave the wrong nucleotide off. This proofreading activity is important
to maintain the high fidelity of the DNA replication process. Mutations, or
sudden changes of nucleotide sequence in DNA, would occur much more
frequently without this activity.
Many sequencing technologies are based on the process of DNA replication.
These technologies, often referred to as sequencing-​by-​synthesis, use this
process to read nucleotide sequence off one strand of the sequencing DNA
target. Corresponding to the components required in the DNA replication
process, these sequencing systems require the following basic components:
(1) sequencing DNA target, which provides the template; (2) nucleotides;
(3) a primer; and (4) a DNA polymerase. Since the DNA polymerase extends
the new strand by attaching one nucleotide at a time, detecting the attached
nucleotide after each extension cycle generates a readout of the nucleo-
tide sequence on the template DNA strand. To facilitate the detection, the
nucleotides used in sequencing reactions are usually chemically modified,
including labeling with fluorescent tags. Chapter 4 focuses on the evolution
of sequencing technologies.
Besides the high fidelity of DNA polymerases, an efficient DNA repair
system is also crucial to maintain genome stability and keep mutation rate low.
Even under normal conditions, DNA nucleotide sequence can be accidently
altered by many physical and chemical factors in the environment, including
intracellularly generated reactive oxygen and nitrogen species, radiation in
the environment (such as UV, X-​ray, or γ-​ray), and other chemical mutagens.
If left uncorrected, these changes will accumulate and cause disturbances to
normal cell function or even cause cell death, leading to diseases. To maintain
the fidelity of DNA molecules, cells invest heavily on DNA repair enzymes.
These enzymes constantly scan genomic DNA and make repairs if damage is
DNA Sequence: The Genome Base 19

DNA Polymerase dNTP


Primer T P P P

5’ A P C P G P A 3’
3’ T P G P C P T P A P G P C P C 5’
Template
P P + H+ Extension
C P P P

5’ A P C P G P A P T 3’
3’ T P G P C P T P A P G P C P C 5’

Extension T P P P
P P + H+

5’ A P C P G P A P T P C 3’

3’ T P G P C P T P A P G P C P C 5’

P P + H+ Mis-incorporation

5’ A P C P G P A P T P C P T 3’

3’ T P G P C P T P A P G P C P C 5’

Error Correction
P T G P P P

5’ A P C P G P A P T P C 3’

3’ T P G P C P T P A P G P C P C 5’

Extension
P P + H+

5’ A P C P G P A P T P C P G 3’
3’ T P G P C P T P A P G P C P C 5’
FIGURE 2.1
The DNA replication process. To initiate the process, a primer, which is a short DNA sequence
complementary to the start region of the DNA template strand, is needed for DNA polymerase
to attach nucleotides and extend the new strand. The attachment of nucleotides is based on
complementary base-​pairing with the template. If an error occurs due to mis-​pairing, the DNA
polymerase removes the mis-​paired nucleotide using its proofreading function. Due to the
biochemical structure of the DNA molecule, the direction of the new strand elongation is from
its 5’ end to 3’ end (the template strand is in the opposite direction; the naming of the two
ends of each DNA strand as 5’ and 3’ is from the numbering of carbon atoms in the nucleotide
sugar ring).
20 Next-Generation Sequencing Data Analysis

detected. The serious consequences of a weakened DNA repair system can be


exemplified by mutations in BRCA2, a gene coding for a DNA repair enzyme,
which lead to breast and ovarian cancers.

2.3 How the Genetic Information Stored in


DNA Is Transferred to Protein
While the logic of the cellular system is written in the nucleotide sequence
of its genomic DNA, almost all cellular activities are executed by the wide
array of proteins in the cell’s proteome. The bio-​information flow from DNA
to protein, known as the central dogma (Figure 2.2), provides a fundamental
framework for modern molecular biology and genetic engineering. Based
on this framework, a gene’s DNA sequence is first transcribed to make
mRNA, and then the nucleotide sequence in mRNA is used to guide the
assembly of amino acids into a protein. The translation of the mRNA nucleo-
tide sequence to the protein amino acid sequence is based on the triplet gen-
etic code. A continuous segment of DNA that contains the full set of triplet
codon for protein translation, from start to stop, is often called an open
reading frame (or ORF). The synthesis of one type of bio-​polymer molecule
based on information stored in another bio-​polymer is one of the greatest
“inventions” of nature.
Since its initial introduction, the central dogma has been gradually modi-
fied with increased sophistication. In its original form, one gene is translated
into one protein via one mRNA. This one gene–​one protein paradigm was
later found to be too simplistic, as one gene can generate multiple forms of

Promoter 3’-UTR
DNA (Gene)

RNA
(Primary Transcript)

mRNA

Exon
Protein
Intron

FIGURE 2.2
The central dogma.
DNA Sequence: The Genome Base 21

proteins through alternative splicing (next chapter). In addition, the informa-


tion flow between DNA and RNA is not simply one-​way from DNA to RNA,
but RNA can also be reverse transcribed to DNA in some organisms. On the
additional role of RNAs in this information flow, some non-​protein-​coding
RNAs can silence gene expression through mechanisms such as inhibiting
gene transcription or translation, or protect genome through mechanisms
like preventing the movement of transposable elements (or transposons,
mobile DNA elements that copy themselves to different genomic loci) (also
see next chapter). Furthermore, chemical modifications of DNA and some
DNA-​interacting proteins constitute the epigenome, which also regulates the
flow of genetic information.

2.4 The Genomic Landscape


2.4.1 The Minimal Genome
After understanding the flow of bio-​information from DNA to protein,
the next question is what is the minimum amount of genetic information
needed to make the cellular system tick, that is, what constitutes the min-
imal genome. Attempts to define the minimal genome started in the late
1950s, shortly after the discovery of the double helix structure of DNA.
The answer to this important question is not straightforward, however,
as the amount of genetic information needed for a minimal life form is
dependent on the specific environment it lives in. Considering the basic
functions that a cell has to perform, the minimal genome needs to con-
tain genes at least for DNA replication, RNA synthesis and processing,
protein translation, energy and molecular metabolism. A small bacterium,
Mycoplasma genitalium, often used as a model of a naturally existing min-
imal genome for a free-​living organism, has a genome of 580 kilobase pairs
(kbp) and 504 genes [1]. An artificially designed and completely synthetic
minimal genome reported in 2016 to support a viable cell contains 531 kbp
sequence and 473 genes [2].

2.4.2 Genome Sizes
For the least sophisticated organisms, such as Mycoplasma genitalium, a min-
imal genome is sufficient. For increased organismal complexity, more genetic
information and, therefore, a larger genome is needed. As a result, there
is a positive correlation between organismal complexity and genome size,
especially in prokaryotes. In eukaryotes, however, this correlation becomes
much weaker, largely due to the existence of non-​coding DNA elements in
varying amounts in different eukaryotic genomes (for details on non-​coding
22 Next-Generation Sequencing Data Analysis

TABLE 2.1
Genome Sizes and Total Gene Numbers in Major Model Organisms* (Ordered by
Genome Size)
Organism Genome Size (bp) Number of Coding Genes

Mycoplasma genitalium (Bacterium) 580,076 504


Haemophilus influenzae (Bacterium) 1,846,259 1,715
Escherichia coli (Bacterium) 4,641,652 4,288
Saccharomyces cerevisiae (Yeast) 12,157,105 6,016
Caenorhabditis elegans (Nematode) 100,286,401 19,984
Arabidopsis thaliana (Thale cress) 119,668,634 27,562
Drosophila melanogaster (Fruit fly) 143,726,002 13,968
Oryza sativa (Rice) 374,422,835 28,738
Medicago truncatula (Legume) 430,008,013 31,927
Danio rerio (Zebrafish) 1,373,454,788 26,522
Zea mays (Maize) 2,182,786,008 34.337
Rattus norvegicus (Rat) 2,647,915,728 22,228
Mus Musculus (Mouse) 2,728,222,451 22,186
Homo sapiens (Human) 3,099,441,038 20,024

*Data based on NCBI genome database as of Nov. 2022.

DNA elements, see Section 2.4.4) and whole-​genome duplication. In terms of


genome size, the currently documented range is 112 kbp, coding 137 genes,
of Nasuia deltocephalinicola (an obligate bacterial endosymbiont living in leaf-
hopper cells) [3] on the lower end, to 149 gigabase pairs (gbp) (number of
genes coded still unknown) of Paris japonica (a slow-​growing flowering plant)
[4] on the higher end. Table 2.1 shows the total number of genes in some of
the most studied organisms.

2.4.3 Protein-​Coding Regions of the Genome


The protein-​coding regions are the part of the genome that we foremost
study and know most about. The content of these regions directly affects pro-
tein synthesis and protein diversity in cells. In prokaryotic cells, function-
ally related protein-​coding genes are often arranged next to each other and
regulated as a single unit known as operon. The gene structure in eukaryotic
cells is more complicated. The coding sequences (CDS) of almost all eukary-
otic genes are not continuous and interspersed among non-​coding sequences.
The non-​coding intervening sequences are called introns (int for intervening),
while the coding regions are called exons (ex for expressed) (see Figure 2.2).
During gene transcription, both exons and introns are transcribed. In the sub-
sequent mRNA maturation process, introns are spliced out and exons are
joined together for protein translation.
In the human genome, the average number of exons per gene is 8.8. The
titin gene, coding for a large abundant protein in striated muscle, has 363
exons, the most in any single gene, and also has the longest single exon
DNA Sequence: The Genome Base 23

(17,106 bp) among all currently known exons. In aggregate the total number
of currently known exons in the human genome is around 180,000. With
a combined size of 30 Mb, they constitute 1% of the human genome.
This collection of all exons in the human genome, or in other eukaryotic
genomes, is termed as the exome. Different from the transcriptome, which
is composed of all actively transcribed mRNAs in a particular sample, the
exome includes all exons contained in a genome. While it only covers a very
small percentage of the genome, the exome represents the most important
and the best annotated part of the genome. Sequencing of the exome has
been used as a popular alternative to whole genome sequencing. While it
lacks on coverage, exome sequencing is more cost effective, faster, and easier
for data interpretation.

2.4.4 Non-​Coding Genomic Elements


While protein-​coding genes are the most studied genomic element, they
may not necessarily be the most abundant part of the genome. Prokaryotic
genomes are usually rich in protein-​coding gene sequences, e.g., they account
for approximately 90% of the E. coli genome. In complex eukaryotic genomes,
however, their percentage is lower. For example, only about 1.5% of the
human genome codes for proteins (Figure 2.3). Among the non-​protein-​
coding sequences in eukaryotic genomes are introns, regulatory sequences,
and other unique non-​coding DNA elements. The regulatory sequences are
genomic elements that are known to regulate gene expression, including
promoters, terminators, enhancers, repressors, and silencers. In comparison,
our current understanding of the other unique non-​coding DNA elements is
the most rudimentary. We know nearly nothing about these elements, with
the exception of non-​coding RNA genes, which include rRNAs, tRNAs, and
other functionally important RNA species, which will be detailed in the next
chapter. As mentioned in Chapter 1, rRNAs are key structural components
of the ribosome and directly involved in protein translation, while tRNAs
transport proper amino acids to the ribosome for protein translation based
on the genetic code.
Repetitive sequences occupy more than half of the human genome, and
are even more pervasive in some other eukaryotic genomes. For example, in
some plants and amphibians, 80% of the genome is composed of repetitive
sequences. The percentage of repetitive sequences in prokaryotic genomes is
relatively lower but still significant. With respect to their internal structures,
some repetitive sequences are tandem repeats, with the basic repeating units
connected head-​to-​tail. In this type of sequence repeats, the length of the
repeating units is highly variable, from <10 bp to thousands of base pairs.
The other major type of sequence repeats are interspersed repeats, present
as a single copy in many genomic loci. These are either transposons, or
retrotransposons, that copy themselves via RNA intermediates. Discovered
by geneticist Barbara McClintock, transposons (also called transposable
24 Next-Generation Sequencing Data Analysis

Exons
(1.5%)

Repetitive
Sequences Not
Related to Introns
Transposons
(20%)
(14%)

Regulatory
Sequences
(5%)

Repetitive
Sequences Other Unique
Related to Non -Coding DNA
Transposons Sequences
(44.5%) (15%)

FIGURE 2.3
The composition of the human genome.

elements, or “jumping genes”) are DNA sequences that move from one gen-
omic location to another. Repeat sequence units of this type are usually 100
bp to over 10 kb in length, and may appear in over 1 million loci dispersed
across the genome.
Many highly repetitive DNA sequences exist in inert parts of chromosomes,
such as the centromere and telomere. The centromere, the region where two
sister chromatids are linked together before cell division, contains tandem
repeat sequences. The telomere, existing at the ends of chromosomes, is
also composed of highly repetitive DNA sequences. The telomeric structure
protects chromosomal integrity and thereby maintains genomic stability.
Besides being essential in maintaining the chromosomal structure, repeat
sequences have other functions in the genome, e.g., they play an architec-
tonic role in higher order physical genome structuring [5]. Despite their
abundance and function, because sequences associated with repeat regions
are not unique, they create a major hurdle for assembling a genome de novo
from sequencing reads, or mapping reads originated from these regions to a
pre-​assembled genome.
DNA Sequence: The Genome Base 25

2.5 DNA Packaging, Sequence Access, and


DNA-​Protein Interactions
2.5.1 DNA Packaging
In the nucleoid of prokaryotic cells, multiple proteins fold and condense gen-
omic DNA into a supercoiled structure to make it fit into the rather limited
space. While being generally condensed, parts of the DNA need to be exposed
to allow sequence access for transcription by related proteins factors. While
these processes have been studied in prokaryotic cells, DNA packaging and
sequence accessing are better studied and understood in eukaryotic cells.
In these cells, because of their much larger genome size, genomic DNA is
condensed in the nucleus to a much higher degree. For instance, the total
length of human genomic DNA is about 2 meters when fully stretched out,
but the diameter of the human cell nucleus is only 6 μm. Bound to specific
proteins called histones, eukaryotic DNA is packaged in the form of chro-
matin, in which the positively charged histones bind to the negatively
charged DNA molecules through electrostatic interactions. This packaging
process involves compacting DNA at different levels. At the first level, DNA
wraps around a protein complex composed of eight histone subunits to form
the basic structure of nucleosome. Each nucleosome contains around 200
nucleotide pairs and has a diameter of 11 nm. At the second level the nucleo-
some structure is compacted into a fiber structure. This fiber, with a diameter
of 30 nm, is the form most chromatin takes in the interphase between two cell
divisions. Prior to cell division, this chromatin fiber is further condensed by
two additional levels into chromosome, the extremely condensed form that
we can observe under a light microscope.

2.5.2 Sequence Access
Since different DNA sequences in the genome are constantly being
transcribed, instead of being permanently locked into the compacted
form, DNA sequences at specific loci need to be dynamically exposed to
allow transcriptional access to protein factors such as transcription factors
and coactivators. Furthermore, DNA replication and repair also require
chromatin unpackaging. This unpackaging of the chromatin structure is
carried out through two principal mechanisms. One is through histone
modification, such as acetylation of lysine residues on histones by histone
acetyltransferases, which reduces the positive charge on histones and there-
fore decreases the electrostatic interactions between histones and DNA.
Deacetylation by histone deacetylases, on the other hand, restricts DNA
access and represses transcription. The other unpackaging mechanism is
through the actions of chromatin remodeling complexes. These large pro-
tein complexes consume ATP and use the released energy to expose DNA
26 Next-Generation Sequencing Data Analysis

sequences for transcription through nucleosomal repositioning, nucleo-


somal eviction, or local unwrapping.

2.5.3 DNA-​Protein Interactions
While DNA is the carrier of the code of life, the DNA code cannot be executed
without DNA-​interacting proteins. Nearly all of the processes mentioned
above, including DNA packaging/​ unpackaging, transcription, repair,
and replication, rely on such proteins. Besides histones, examples of these
proteins include transcription factors, RNA polymerases, DNA polymerases,
and nucleases (for DNA degradation). Many of these proteins, such as
histones and DNA/​RNA polymerases, interact with DNA regardless of their
sequence or structure. Some DNA-​interacting proteins bind to DNA of spe-
cial structure/​conformation, e.g., high-​mobility group (HMG) proteins that
have high affinity for bent or distorted DNA. Some other DNA-​interacting
proteins bind only to regions of the genome that have certain characteristics
such as having damage, the examples of which are DNA repair enzymes such
as BRCA1, BRCA2, RAD51, RAD52, and TDG.
The most widely studied DNA-​ interacting proteins are transcription
factors, which bind to specific DNA sequences. Through binding to their
specific recognition sequences in the genome, transcription factors regu-
late transcription of gene targets that contain such sequences in their pro-
moter region. Since they bind to more than one gene locations in the genome,
transcription factors regulate the transcription of a multitude of genes in
a coordinated fashion, usually as a response to certain internal or external
environmental change. For instance, NRF2 is a transcription factor that is
activated in response to oxidative stress. Upon activation, it binds to a short
segment of specific DNA sequence called the anti-​oxidant response element
(ARE) located in the promoter region of those genes that are responsive to
oxidative stress. Through binding to this sequence element in many regions
of the genome, NRF2 regulates the transcription of its target genes and
thereby elicits coordinated responses to counteract the damaging effects of
oxidative stress.
Study of DNA-​ protein interactions provides insights into how the
genome responds to various conditions. For example, determination of
transcription factor binding sites, such as those of NRF2, across the genome
can unravel what genes might be responsive to the conditions that activate
the transcription factors. While such sites can be predicted computationally,
only wet-​lab experiment can determine where a transcription factor actu-
ally binds in the genome under a certain condition. ChIP-​seq, or chromatin
immunoprecipitation coupled with sequencing, is one application of NGS
that is developed to study genomic binding of transcription factors and
other DNA-​interacting proteins. Chapter 13 will focus on ChIP-​seq data
analysis.
DNA Sequence: The Genome Base 27

2.6 DNA Sequence Mutation and Polymorphism


While DNA replication is a high-​fidelity process and the nucleus maintains
an army of DNA repair enzymes, sequence mutation does happen, although
at a very low frequency. In general, the rate of mutation in prokaryotic and
eukaryotic cells is at the scale of 10−​9 per base per cell division. In multi-​
cellular eukaryotic organisms, germline cells have lower mutation rate than
somatic cells. In these organisms, because most cells, including germline
cells, undergo multiple divisions in the organisms’ lifetime, the per-​
generation mutation rate is significantly higher. For example, whole genome
sequencing data collected from human blood cell DNA estimates a mutation
rate of 1.1 × 10−​8 per base per generation, corresponding to around 70 new
mutations in each human diploid genome [6]. Depending on the nature of the
change, mutations may have deleterious, neutral, or rarely, beneficial effects
on the organism. Mutations lead to sequence variation, and are ultimately
the basis of genome evolution and diversification for those carried through
the germline. Although mutations in somatic cells are not passed on to the
next generation, they can lead to diseases, including cancer, and affect the
survival of the individual.
There are various forms of DNA mutations, from single nucleotide
substitutions, small insertions/​deletions (or indels), to structural variations
(SVs) that involve larger genomic regions. Among these different types of
mutations, single nucleotide substitutions, also called point mutations, are the
most common. These substitutions can be either transitions or transversions.
Transitions involve the substitution of a purine for the other purine (i.e.,
A↔G) or a pyrimidine for the other pyrimidine (i.e., C↔T). Transversions,
on the other hand, involve the substitution of a purine for a pyrimidine, or
vice versa. Theoretically there are more combinations of transversions than
transitions, but due to the nature of the underlying biochemical processes
transitions actually occur more frequently than transversions. If single
nucleotide substitution takes place in a protein-​coding region, it may or
may not lead to change in amino acid coding. If it causes the substitution
of one amino acid for another, it is a missense mutation, which may lead to
change of protein function. If it introduces a stop codon and as a result leads
to the generation of a truncated protein, it is a nonsense mutation. Both the
missense and nonsense mutations are non-​synonymous mutations. If it does
not change the coded amino acid due to the redundancy in the genetic code,
it is a synonymous mutation and has no effect on protein function. Because
of its common occurrence, single nucleotide variation (SNV) is the most fre-
quently observed sequence variation. If an SNV is commonly observed in
a population, it is called a single nucleotide polymorphism (or SNP). More
than 1,000 million SNPs in the human genome have been cataloged at the
time of this writing (November 2022). Because of their high density in the
28 Next-Generation Sequencing Data Analysis

genome, SNPs are often used as flagging markers to cover the entire genome
in high resolution when scanning for genomic region(s) that are associated
with a phenotype or disease of interest.
Besides single nucleotide substitutions, indels are another common type
of mutation. Most indels involve small numbers of nucleotides. In protein-​
coding regions, small indels lead to the shift of ORF (unless the number of
nucleotides involved is a multiple of three), resulting in the formation of a
vastly different protein product. Indels that involve large regions lead to
alterations of genomic structure and are usually considered as a form of SV.
Besides large indels, SVs, defined as changes encompassing at least 50 bp [7],
also include inversions, translocations, or duplications. Copy number vari-
ation (CNV) is a subcategory of SV, usually caused by large indel or seg-
mental duplication. Although they affect larger genomic region(s) and some
lead to observable phenotypic changes or diseases, many CNVs, or SVs in
general, have no detectable effects. The frequency of SVs in the genome was
underestimated previously due to technological limitations. The emergence
of NGS has greatly enabled SV detection, which has led to the realization of
its wide existence [8].

2.7 Genome Evolution
The spontaneous mutations that lead to sequence variation and poly-
morphism in a population are also the fundamental force behind the evo-
lution of genomes and eventually the Darwinian evolution of the host
organisms. Gradual sequence change and diversification of early genomes,
over billions of years, have evolved into the extremely large number of
genomes that had existed or are functioning in varying complexity today. In
this process, existing DNA sequences are constantly modified, duplicated,
and reshuffled. Most mutations in protein-​coding or regulatory sequences
disrupt the protein’s normal function or alter its amount in cells, causing cel-
lular dysfunction and affecting organismal survival. Under rare conditions,
however, a mutation can improve existing protein function or lead to the
emergence of new functions. If such a mutation offers its host a competitive
advantage, it is more likely to be selected and passed on to future generations.
Gene duplication provides another major mechanism for genome evolu-
tion. If a genomic region containing one or multiple gene(s) is duplicated
resulting in the formation of an SV, the duplicated region is not under selec-
tion pressure and therefore becomes substrate for sequence divergence and
new gene formation. Although there are other ways of adding new genetic
information to a genome such as inter-​species gene transfer, DNA duplica-
tion is believed to be a major source of new genetic information generation.
DNA Sequence: The Genome Base 29

Gene duplication often leads to the formation of gene families. Genes in the
same family are homologous, but each member has their specific function and
expression pattern. As an example, in the human genome there are 339 genes
in the olfactory receptor gene family. Odor perception starts with the binding
of odorant molecules to olfactory receptors located on olfactory neurons
inside the nose epithelium. To detect different odorants, combination of
different olfactory receptors that are coded by genes in this family is required.
Based on their sequence homology, members of this large family can be even
further grouped into different subfamilies [9]. The existence of pseudogenes
in the genome is another result of gene duplication. After duplication, some
genes may lose their function and become inactive from additional mutation.
Pseudogenes may also be formed in the absence of duplication by the disab-
ling of a functional gene from mutation. A pseudogene called GULO mapped
to the human chromosome 8p21 provides such an example. The functional
GULO gene in other organisms codes for an enzyme that catalyzes the last
step of ascorbic acid (vitamin C) biosynthesis. This gene is knocked out in
primates, including human, and becomes a pseudogene. As a result, we have
to get this essential vitamin from food. The inactivation of this gene is pos-
sibly due to the insertion to the gene’s coding sequence of a retrotransposon-​
type repetitive sequence called Alu element [10].
DNA recombination, or reshuffling of DNA sequences, also plays an
important role in genome evolution. Although it does not create new genetic
information, by breaking existing DNA sequences and re-​joining them DNA
recombination changes the linkage relationships between different genes
and other important regulatory sequences. Without recombination, once a
harmful mutation is formed in a gene, the mutated gene will be permanently
linked to other nearby functional genes, and impossible to regroup all the
functional genes back together into the same DNA molecule. Through this
regrouping, DNA recombination makes it possible to avoid gradual accumu-
lation of harmful gene mutations. Most DNA recombination events happen
during meiosis in the formation of gametes (sperm or eggs) as part of sexual
reproduction.

2.8 Epigenome and DNA Methylation


Besides the regulatory DNA sequences introduced earlier, chemical
modifications of specific nucleotides in the genome, like the acetylation and
deacetylation of histones, offer another layer of regulation on genetic activ-
ities. Since they provide additional genetic activity regulation, these chemical
modifications on DNA and histones constitute the epigenome. Methylation
of the fifth carbon on cytosine (5-​methylcytosine, or 5mC) is currently the
most studied epigenomic modification in many organisms. Enzymatically,
30 Next-Generation Sequencing Data Analysis

this methylation is carried out and maintained by DNA methyltransferases


(three identified in mammals: DNMT1, DNMT3A, and DNMT3B). The
cytosines that undergo methylation can occur in three different sequence
contexts –​CpG, CHG, and CHH (H can be A, G, or T) –​each involving
different pathways [11]. Most methylated cytosines exist in the CpG con-
text, where the methylation reduces gene expression through recruiting gene
silencing proteins, or preventing transcription factors from binding to the
DNA. The methylation of cytosines in this context also affects nucleosome
positioning and chromatin remodeling, as methyl-​ CpG-binding domain
(MBD) proteins that specifically bind to 5mC at CpG sites can recruit histone
modifying proteins and those in the chromatin remodeling complex [12]. The
effects of cytosine methylation in the CHG and CHH contexts are less clear
but currently available data seems to suggest that they may play a regulatory
role in repetitive regions [13].
Just like deacetylation counteracts the effects of acetylation in histones,
demethylation of cytosines should be similarly important to reverse the
effects of 5mC when the methylation is no longer needed. Until recently,
the steps involved in the cytosine demethylation process began to be under-
stood. In this process, the 5mC is first oxidized to 5-​hydroxymethylcytosine
(5hmC), and then to 5-​formylcytosine (5fC) and 5-​carboxylcytosine (5caC)
in mammals. These oxidative conversions are catalyzed by enzyme systems
such as the TET family proteins. The subsequent base excision repair of 5fC/​
5caC by an enzyme called TDG, or 5mC directly by the glycosylase enzyme
in plants, completes the DNA demethylation process [14]. Compared to 5mC,
the levels of these demethylation intermediate products are detected to be
much lower in most cells (except that 5hmC has been found to be relatively
abundant in embryonic stem cells and in the brain).
Different from the genome, which is static, the epigenome is dynamic
and changes with environmental conditions. These dynamically chan-
ging epigenomic modifications regulate gene expression and thereby play
important roles in embryonic development, cell differentiation, stem cell
pluripotency, genomic imprinting, and genome stability. In accordance with
their regulatory functions, these modifications are highly site specific. To
study where cytosine methylations take place in the genome, multiple NGS-​
based approaches, which will be detailed in Chapter 14, have been developed
and widely applied to epigenomics studies. Methodological development
for the study of cytosine demethylation is currently still at an early stage.

2.9 Genome Sequencing and Disease Risk


The wide accessibility of DNA sequences, largely fueled by the rapid devel-
opment of new sequencing technologies, has uncovered extensive sequence
DNA Sequence: The Genome Base 31

variation in individual genomes within a population. The extensiveness in


sequence variation was not envisioned in early days of genetics, not even
when the Human Genome Project was completed in 2003. This has grad-
ually led to a paradigm shift in disease diagnosis and prevention. As a result,
the public becomes more aware of the role of individual genomic makeup
in disease development and predisposition. In addition, the easier accessi-
bility to our DNA sequence has further prompted us to look into our genome
and use that information for preemptive disease prevention. The declining
cost of genome sequencing has also enabled the biomedical community to
dig deeper into the genomic underpinnings of diseases, by unraveling the
linkage between sequence polymorphism in the genome and disease inci-
dence. Below is a brief overview of the major categories of human diseases
that have an intimate connection with DNA mutation, polymorphism,
genome structure, and epigenomic abnormality.

2.9.1 Mendelian (Single-​Gene) Diseases


The simplest form of hereditary diseases is caused by mutation(s) in a
single gene, and therefore also called monogenic or Mendelian diseases. For
example, sickle-​cell anemia is caused by a mutation in the HBB gene located
on the human chromosome 11. This gene codes for the β subunit of hemo-
globin, an important oxygen-​carrying protein in the blood. A mutation of this
gene leads to the replacement of the sixth amino acid, glutamic acid, with
another amino acid valine in the coded protein. This change of a single amino
acid causes conformational change of the protein, leading to the generation
of sickle-​shaped blood cells that die prematurely. This disease is recessive,
meaning that it only appears when both copies (or alleles) of the gene carry
the mutation. In dominant diseases, however, one mutant allele is enough to
cause sickness. Huntington’s disease, a neurodegenerative disease that leads
to gradual loss of movement control and mental faculties, is such a dominant
single-​gene disease. It is caused by mutation in a gene called HTT on the
human chromosome 4, coding for a protein called huntingtin. The involved
mutation is an expansion of tri-​nucleotide (CAG, codes for the amino acid
glutamine) repeats. When the number of CAG repeats is higher than 36, it
leads to the production of an abnormally long polyglutamine tract in the
huntingtin protein. This confers a dominant deleterious gain of function on
the protein, causing neuronal damage and eventually loss in the striatum and
cortical regions of the brain.

2.9.2 Complex Diseases That Involve Multiple Genes


Most common diseases, including heart disease, diabetes, hypertension,
obesity, and Alzheimer’s disease (AD), are caused by multiple genes. In the
case of AD, while its familial or early-​onset form can be attributed to one
of three genes (APP, PSEN1, and PSEN2), the most common form, sporadic
32 Next-Generation Sequencing Data Analysis

AD, involves a large number of genes [15]. In this type of complex disease,
the contribution of each gene is modest, and it is the combined effects of
mutations in these genes that predispose an individual to these diseases.
Besides genetic factors, lifestyle and environmental factors often also play
a role. For example, history of head trauma, lack of mentally stimulating
activities, and high cholesterol levels are all risk factors for developing AD.
Because of the number of genes involved and their interactions with non-​
genetic factors, complex multi-​gene diseases are more challenging to study
than single-​gene diseases.

2.9.3 Diseases Caused by Genome Instability


Aside from the gene-​centered disease models introduced above, diseases can
also occur as consequences of large-​scale genomic changes such as rearrange-
ment of large genomic regions, alterations of chromosome number, and gen-
eral genome instability. For example, when a genome becomes unstable in an
organism, it can cause congenital developmental defects, tumorigenesis, pre-
mature aging, etc. Dysfunction in genome maintenance, such as DNA repair
and chromosome segregation, can lead to genome instability. Fanconi anemia
is an example of a disease caused by genome instability, characterized by
growth retardation, congenital malformation, bone marrow failure, high
cancer risk, and premature aging. The genome instability in this disease is
caused by mutations in a cluster of DNA repair genes, and manifested by
increased mutation rates, cell cycle disturbance, chromosomal breakage, and
extreme sensitivity to reactive oxygen species and other DNA damaging
agents.
Cancer, to a large degree, is also caused by genome instability. This can
be hinted by the fact that two well-​known high-​risk cancer genes, BRCA1
and BRCA2, are both DNA damage repair genes. Mutations in the two
genes greatly increase the susceptibility to tumorigenesis, such as breast and
ovarian cancers. In general, many cancers are characterized by chromosomal
aberrations and genome structural changes, involving deletion, duplication,
and rearrangement of large genomic regions. The fact that genome instability
is intimately related to major aspects of cancer cells such as cell cycle regu-
lation and DNA damage repair also points to the important role of genome
instability in cancer development.

2.9.4 Epigenomic/​Epigenetic Diseases
Besides gene mutations and genome instability, abnormal epigenomic/​epi-
genetic pattern can also lead to diseases. Examples of diseases in this cat-
egory include fragile X syndrome, ICF (immunodeficiency, centromeric
instability and facial anomalies) syndrome, Rett syndrome, and Rubinstein-​
Taybi syndrome. In ICF syndrome, for example, the gene DNMT3B is
DNA Sequence: The Genome Base 33

mutated leading to the deficiency of DNA methyltransferase 3B. As a result,


patients afflicted with this disease invariably have DNA hypomethylation.
Cancer, as a genome disease that is caused by more than one genetic/​gen-
omic factor, is also characterized by abnormal DNA methylation, including
both hypermethylation and hypomethylation. The hypermethylation is com-
monly observed in the promoter CpG islands of tumor suppressor genes
[16], which leads to their suppressed transcription. The hypomethylation
is mostly located in highly repetitive sequences, including tandem repeats
in the centromere and interspersed repeats. This lowered DNA methylation
has been suggested to play a role in promoting chromosomal relaxation and
genome instability [17].

References
1. Fraser CM, Gocayne JD, White O, Adams MD, Clayton RA, Fleischmann RD,
Bult CJ, Kerlavage AR, Sutton G, Kelley JM et al. The minimal gene comple-
ment of Mycoplasma genitalium. Science 1995, 270(5235):397–​403.
2. Hutchison CA, 3rd, Chuang RY, Noskov VN, Assad-​Garcia N, Deerinck TJ,
Ellisman MH, Gill J, Kannan K, Karas BJ, Ma L et al. Design and synthesis of a
minimal bacterial genome. Science 2016, 351(6280):aad6253.
3. Bennett GM, Moran NA. Small, smaller, smallest: the origins and evolution
of ancient dual symbioses in a Phloem-​feeding insect. Genome Biol Evol 2013,
5(9):1675–​1688.
4. Pellicer J, Fay MF, Leitch IJ. The largest eukaryotic genome of them all? Bot J
Linn Soc 2010, 164(1):10–​15.
5. Shapiro JA, von Sternberg R. Why repetitive DNA is essential to genome
function. Biol Rev Camb Philos Soc 2005, 80(2):227–​250.
6. Roach JC, Glusman G, Smit AF, Huff CD, Hubley R, Shannon PT, Rowen
L, Pant KP, Goodman N, Bamshad M et al. Analysis of genetic inher-
itance in a family quartet by whole-​ genome sequencing. Science 2010,
328(5978):636–​639.
7. Mahmoud M, Gobet N, Cruz-​Davalos DI, Mounier N, Dessimoz C, Sedlazeck
FJ. Structural variant calling: the long and the short of it. Genome Biol 2019,
20(1):246.
8. Ebert P, Audano PA, Zhu Q, Rodriguez-​Martin B, Porubsky D, Bonder MJ,
Sulovari A, Ebler J, Zhou W, Serra Mari R et al. Haplotype-​resolved diverse
human genomes and integrated analysis of structural variation. Science 2021,
372(6537):eabf7117.
9. Malnic B, Godfrey PA, Buck LB. The human olfactory receptor gene family.
Proc Natl Acad Sci U S A 2004, 101(8):2584–​2589.
10. Inai Y, Ohta Y, Nishikimi M. The whole structure of the human nonfunctional
L-​gulono-​gamma-​lactone oxidase gene—the gene responsible for scurvy—​
and the evolution of repetitive sequences thereon. J Nutr Sci Vitaminol 2003,
49(5):315–​319.
34 Next-Generation Sequencing Data Analysis

11. Law JA, Jacobsen SE. Establishing, maintaining and modifying DNA methyla-
tion patterns in plants and animals. Nat Rev Genet 2010, 11(3):204–​220.
12. Cedar H, Bergman Y. Linking DNA methylation and histone modifica-
tion: patterns and paradigms. Nat Rev Genet 2009, 10(5):295–​304.
13. Guo W, Chung WY, Qian M, Pellegrini M, Zhang MQ. Characterizing the
strand-​specific distribution of non-​CpG methylation in human pluripotent
cells. Nucleic Acids Res 2014, 42(5):3009–​3016.
14. Wu H, Zhang Y. Reversing DNA methylation: mechanisms, genomics, and
biological functions. Cell 2014, 156(1–​2):45–​68.
15. Shademan B, Biray Avci C, Nikanfar M, Nourazarian A. Application of next-​
generation sequencing in neurodegenerative diseases: opportunities and
challenges. Neuromolecular Med 2021, 23(2):225–​235.
16. Nishiyama A, Nakanishi M. Navigating the DNA methylation landscape of
cancer. Trends Genet 2021, 37(11):1012–​1027.
17. Pappalardo XG, Barra V. Losing DNA methylation at repetitive elements and
breaking bad. Epigenetics Chromatin 2021, 14(1):25.
3 
RNA: The Transcribed Sequence

3.1 RNA as the Messenger


The blueprint of life is written in DNA, but almost all life processes are
executed by proteins. To convert the information coded in the DNA into the
wide array of proteins in each cell, segments of DNA sequence in the genome
must be copied into mRNAs first. The transcribed nucleotide sequences in the
mRNAs are then translated into proteins through an information decoding
process carried out by ribosomes. Because of the intermediary role played
by mRNAs between DNA and proteins, the composition of mRNAs in a cell
or population of cells –​the transcriptome –​is often used to study cellular
processes and functions. Unlike the genome, which is static and the same for
every cell in an organism, the transcriptome is dynamically regulated and
therefore can be used as a proxy of cellular functional status.

3.2 The Molecular Structure of RNA


Structurally RNA is closely related to DNA and also made of nucleotides. The
nucleotides that make up the RNA molecule are slightly different from those
of DNA. Instead of deoxyribose, its five-​carbon sugar moiety is a ribose.
Among the four nucleobases, uracil (U) is used in place of thymine (T), while
the rest of three (A, C, and G) are the same. Unlike the double-​stranded struc-
ture of DNA, RNA molecules are single-​stranded, which gives them great
flexibility. If intramolecular sequence complementarity exists between two
regions of a single RNA molecule, this structural flexibility allows the regions
to bend back on each other and form intramolecular interactions.
As a result of its structural flexibility and internal sequence comple-
mentarity, an RNA molecule can assume secondary structures, such as
hairpins and stem-​loops, and tertiary structures depending on its specific

DOI: 10.1201/9780429329180-4 35
36 Next-Generation Sequencing Data Analysis

sequence. These structures can sometimes afford them special chemical


properties in cells. For example, some non-​messenger RNAs can catalyze
chemical reactions like protein enzymes, and are therefore called RNA
enzymes (or ribozymes, more details later in Section 3.4.1). Some RNA
molecules may assume tertiary structures that enable them to bind to other
small molecules such as ligands, or large molecules such as RNA-​binding
proteins. For mRNAs, their structures may also be important for various
steps of their life cycle (see next section for details). One example of this is
riboswitch, a region in some mRNAs that binds to small molecule ligands
such as metabolites or ions, and thereby regulates their transcription, trans-
lation, or splicing via changes in RNA structure upon ligand binding [1].
Binding of proteins to mRNA elements like those located in the 3’ untrans-
lated region (UTR) can also induce structural changes of these elements
and affect mRNA translation [2]. Transport of mRNAs to specific cellular
locations, such as distal dendritic regions of a neuron, also requires the
mRNAs to assume specific structures for RNA-​binding proteins to bind as
a prerequisite of the transport process. To study structures of individual
RNAs, computational prediction and experimental approaches such as RNA
fingerprinting that uses a variety of chemical and enzymatic probes, have
been the classic methods. With the advent of NGS-​based RNA sequencing,
transcriptome-​wide RNA structural mapping is enabled when integrated
with these classic approaches [3].

3.3 Generation, Processing, and Turnover


of RNA as a Messenger
When a protein is needed in a cell, its coding gene is first transcribed to mRNA,
which is then used as the template to translate into the requisite protein. In
a prokaryotic cell, mRNA transcription is immediately followed by protein
translation. In a eukaryotic cell, the information flow from DNA to protein
through mRNA is more complex, because the two steps of transcription and
translation are physically separated and eukaryotic genes contain introns
that need to be first removed before translation. In the eukaryotic system, ini-
tial transcript (also called primary transcript) is first synthesized from DNA
template and then processed, including intron removal, to produce mature
mRNA in the nucleus. Then the mRNA is transported from the nucleus to
the cytoplasm for translation. When they are no longer needed, the mRNAs
are degraded and recycled by the cell. It should also be noted that the tran-
scription process generates a number of mRNA copies from a gene, and the
number of copies varies from condition to condition and from gene to gene
depending on cellular functional status.
RNA: The Transcribed Sequence 37

FIGURE 3.1
How the two strands of DNA template match the transcribed mRNA in sequence, and the
genetic code in mRNA sequence corresponds to peptide amino acid sequence.

3.3.1 DNA Template
To initiate transcription, a gene’s DNA sequence is first exposed through
altering its packing state. In order to transcribe the DNA sequence, the two
DNA strands in the region are first unwound and only one strand is used as
the template strand for transcription. Since it is complementary to the RNA
transcript in base pairing (A, C, G, and T in the DNA template are transcribed
to U, G, C, and A, respectively, in the RNA transcript), this DNA template
strand is also called the antisense or negative (–​) strand (Figure 3.1). The
other DNA strand has the same sequence as the mRNA (except with T’s in
DNA being replaced with U’s in RNA) and is called the coding strand, sense,
or positive (+​) strand. It should be noted that either strand of the genomic
DNA can be potentially used as the template, and which strand is used as the
template for a gene depends on the orientation of the gene along the DNA. It
should also be noted that the triplet nucleotide genetic code that determines
how amino acids are assembled in proteins refers to the triplet sequence in
the mRNA sequence.

3.3.2 Transcription of Prokaryotic Genes


RNA polymerase catalyzes the transcription of RNA from its DNA template.
In prokaryotic cells, there is only one type of RNA polymerase. The prokary-
otic RNA polymerase holoenzyme contains a core enzyme of five subunits
that catalyzes RNA transcription from a DNA template, and another sub-
unit called the sigma factor that is required for initiation of transcription.
The sigma factor initiates the process by enabling binding of the core enzyme
to the promoter region and guiding it to the transcription start site (TSS).
Promoter is the region upstream of the protein-​coding sequence of a gene or
an operon. Prokaryotic promoters share some core sequence elements, such
as the motif centered at 10 nucleotides upstream of TSS with the consensus
sequence TATAAT. Once reaching TSS, the sigma factor disassociates from
38 Next-Generation Sequencing Data Analysis

the core enzyme. The core RNA polymerase, unlike DNA polymerase, does
not need a primer, but otherwise the enzyme catalyzes the attachment of
nucleotides to the nascent RNA molecule one at a time in the 5’→3’ direction.
At a speed of approximately 30 nucleotides/​second, the RNA polymerase
slides through the DNA template carrying the elongating RNA molecule.
Although the attachment of new nucleotides to the elongating RNA is
based on base pairing with the DNA template, the new elongating RNA
does not remain associated with the template DNA via hydrogen bonding.
On the same template multiple copies of RNA transcripts can be simul-
taneously synthesized by multiple RNA polymerases one after another.
During transcript elongation, these polymerases hold onto the tem-
plate tightly and do not disassociate from the template until stop signal
is transcribed. The stop signal is provided by a segment of palindromic
sequence located at the end of the transcribed sequence. Right after tran-
scription, the inherent self-​complementarity in the palindromic sequence
leads to the spontaneous formation of a hairpin structure. Additional stop
signal is also provided by a string of four or more uracil residues after the
hairpin structure, which forms weak associations with the complementary
A’s on the DNA template. The hairpin structure pauses further elongation
of the transcript, and the weak associations between the U’s on the RNA
and the A’s on the DNA dissociate the enzyme and the transcript from the
template.
Regulation of prokaryotic transcription is conferred by promoters and
protein factors such as repressors and activators. Promoter strength, that
is, the number of transcription events initiated per unit time, varies widely
in different operons. For example, in E. coli, genes in operons with weak
promoters can be transcribed once in 10 minutes, while those with strong
promoters can be transcribed 300 times in the same amount of time. The
strength of an operon’s promoter is based on the host cell’s demand for its
protein products, and dictated by its sequence. Specific protein factors may
also regulate gene transcription. Repressors, the best known among these
factors, prevents RNA polymerase from initiating transcription through
binding to an intervening sequence between promoter and TSS called oper-
ator. Activators exert an opposite effect and induce higher levels of transcrip-
tion. The sigma factor, being the initiation factor of the prokaryotic RNA
polymerase, provides another mechanism for regulation. There are different
forms of this factor in prokaryotic cells, each of which mediates sequence spe-
cific transcription. Differential use of these sigma factors, therefore, provides
another level of transcriptional regulation in prokaryotic cells.

3.3.3 Pre-​mRNA Transcription of Eukaryotic Genes


In eukaryotic cells there are three types of RNA polymerases, among
which RNA polymerase II transcribes protein-​coding genes, while RNA
polymerases I and III transcribe rRNA, tRNA, and various types of small
RNA: The Transcribed Sequence 39

RNAs. Transcription in eukaryotic cells is in general much more sophisticated,


because of the highly compressed packaging of chromosomal DNA, the
complex structure of eukaryotic genes, and intricate regulation by multiple
factors. Prior to transcription, the highly compressed DNA in the chromatin
needs to be uncompressed and gene sequence exposed for access by RNA
polymerase.
To perform the transcription of protein-​coding genes, besides RNA poly-
merase II, a variety of other proteins in the nucleus are also required, including
transcription factors and coactivators. Transcription factors include general
and specific transcription factors. General transcription factors, such as TFIIA,
TFIIB, and TFIID, are required in all transcription initiation. Their functions
are to position the RNA polymerase at the promoter region and unwind the
template DNA strands for transcription. Specific transcription factors, which
are detailed next, provide key regulatory function to the transcription initi-
ation process. Coactivators bring together all requisite transcription factors
to form the transcription initiation complex. Once transcription is initiated,
most of the protein factors in the complex are released and the RNA elong-
ation process is carried out by RNA polymerase II in a manner similar to
that occurs in prokaryotic cells. The termination of the elongation process
in eukaryotic cells is provided by the signal sequence AAUAAA, which also
serves as the signal for cleavage of the transcribed RNA to generate the 3’ end
and for polyadenylation (see Section 3.3.4). After completion of the transcrip-
tion process, the transcript contains both exons and introns and is called the
primary transcript or pre-​mRNA.
During RNA transcript elongation in both the eukaryotic and prokaryotic
systems, like in DNA replication by the DNA polymerase, there is a certain
probability of introducing mismatched nucleotides and therefore errors. For
proofreading, the prokaryotic and eukaryotic RNA polymerases have 3’→5’
exonuclease activity. If a wrong nucleotide is added to the elongating RNA
chain, the RNA polymerase will backtrack and correct the error. Because
of this activity, the overall error rate of the transcriptional process in both
systems is estimated to be 10−​4-​10−​5 per base [4]. Although this is higher
than the DNA mutation rate, the transcriptional errors are seldom harmful,
because there are multiple copies for each transcript, and transcripts carrying
premature stop codons are quickly removed by a process called nonsense-​
mediated decay.
Besides the step of gene sequence exposure through histone modifications
and chromatin remodeling, the eukaryotic gene transcription process is
mostly regulated at the initiation step through the use of specific transcrip-
tion factors. As a large group of DNA-​interacting proteins (Chapter 2), these
transcription factors bind to specific sequence elements in the promoter
region of genes, through which they help assemble general transcription
factors and the RNA polymerase into the transcription initiation complex.
In addition, specific transcription factors may also bind to specific regula-
tory sequences at distant locations that are called enhancers or cis-​regulatory
40 Next-Generation Sequencing Data Analysis

modules. Different from transcription factor binding sites in the promoter


regions, enhancers function independent of sequence orientation and from
a distance as far as megabases away from the regulated gene, and are
sometimes embedded in intergenic regions that otherwise have no known
function. Having a significant effect on gene transcription, enhancers exert
their regulatory function by DNA looping, which brings enhancer and pro-
moter sequences together affecting formation of the transcription initiation
complex. The binding of specific transcription factors to enhancers can have
a stimulatory, or inhibitory (through the recruitment of repressors), effect on
gene transcription. In general, the transcription of a gene is often regulated
by multiple specific transcription factors, and the combined signal input from
these transcription factors determines whether the gene will be transcribed,
and if yes, at what level. A particular transcription factor can also bind to
multiple genomic sites, coordinating the transcription of functionally related
genes. NGS-​based approaches, such as ChIP-​seq (Chapter 13), are often used
to locate the binding sites of specific transcription factors across the genome.

3.3.4 Maturation of mRNA
In prokaryotic cells, there is no post-​ transcription RNA processing, and
transcripts are immediately ready for protein translation after transcription.
In fact, while mRNAs are still being transcribed, ribosomes already bind to
the transcribed portions of the elongating mRNAs synthesizing peptides. In
eukaryotic cells, however, primary transcripts undergo several steps of pro-
cessing in the nucleus to become mature mRNAs. These steps are (1) capping
at the 5’ end, (2) splicing of exons and introns, and (3) addition of a poly-​A
tail at the 3’ end.
The first step, adding a methylated guanosine triphosphate cap to the 5’
end of nascent pre-​mRNAs, takes place shortly after the initiation of tran-
scription when the RNA chains are still less than 30 nucleotides long. This
step is carried out by adding a guanine group to the 5’ end of the transcripts,
followed by methylation of the group. This cap structure marks the transcripts
for subsequent transport to the cytoplasm, protects them from degradation,
and promotes efficient initiation of protein translation. Once formed, the cap
is bound by a protein complex called cap-​binding complex.
The second step, splicing of exons and introns, is the most complicated
among the three steps. As introns are non-​coding intervening sequences,
they need to be spliced out while exons are retained to generate mature
mRNAs. The molecular machinery that carries out the splicing, called the
spliceosome, is assembled from as many as 300 proteins and 5 small nuclear
RNAs (snRNAs). The spliceosome identifies and removes introns from pri-
mary transcripts, using three positions within each intron: the 5’ end (starts
with the consensus sequence 5’-​GU, serving as the splice donor), the 3’ end
(ends with the consensus sequence AG-​3’, as the splice acceptor), and the
branch point, which starts around 30 nucleotides upstream of the splicing
RNA: The Transcribed Sequence 41

acceptor and contains an AU-​rich region. The actual excision of each intron
and the concomitant joining of the two neighboring exons are a three-​step
process: (1) cleavage at the 5’ end splice donor site; (2) attachment of the
cleaved splice donor site to the branch point to form a lariat or loop structure;
and (3) cleavage at the 3’ end splicing acceptor site to release the intron and
join the two exons.
Beyond simply removing introns from primary transcripts, the splicing
process also employs differential use of exons, and sometimes even includes
some introns, to create multiple mature mRNA forms from the same primary
transcript. This differential splicing, also called alternative splicing
(Figure 3.2), provides an additional regulatory step in the production of
mRNA populations. When it was first reported in 1980, alternative splicing
was considered to be an exception rather than the norm. It is now well
established that primary transcripts from essentially all multi-​exon genes are
alternatively spliced [5, 6]. The biological significance of alternative splicing
is obvious: by enabling production of multiple mRNAs and thereby proteins
from the same gene, it greatly augments protein and consequently functional
diversity in an organism without significantly increasing the number of genes
in the genome, and offers explanation to the question why more evolved
organisms do not contain many more genes in their genomes (Chapter 2,
Table 2.1).

Exon Skipping

Use of Mutually Exclusive Exons

Intron Retention

Use of Alternative Promoters

Use of Alternative Polyadenylation Signals

FIGURE 3.2
Varying forms of RNA transcript splicing.
42 Next-Generation Sequencing Data Analysis

In the third step, once the new primary transcript passes the termination
signal sequence, it is bound by several termination-​related proteins. One of
the proteins cleaves the RNA at a short distance downstream of the termin-
ation signal to generate the 3’ end. This is followed by a polyadenylation step
that adds 50–​200 A’s to the 3’ end by an enzyme called poly-​A polymerase.
This poly-​A tail, like the 5’ end cap, increases the stability of the resulting
mRNA. This tail is bound and protected by poly(A)-​binding protein, which
also promotes its transport to the cytoplasm.
Besides these three major constitutive processing steps, some transcripts
may undergo additional processing steps. RNA editing, although considered
to be rare, is among the best known of these steps. RNA editing refers to
the change in RNA nucleotide sequence after it is transcribed. The most
common types of RNA editing are conversions from A to I (inosine, read as G
during translation), which are catalyzed by enzymes such as ADARs (adeno-
sine deaminases that act on RNA), or from C to U, catalyzed by cytidine
deaminases. As a result of these conversions, an edited RNA transcript no
longer fully matches the sequence on the template DNA. RNA editing has the
potential to change genetic codons, introduce new or remove existing stop
codons, or alter splicing sites [7]. Evidence shows that RNA editing and other
RNA processing events such as splicing can be coordinated [8].

3.3.5 Transport and Localization


After maturation, mRNAs need to be exported out of the nucleus to the cyto-
plasm for protein translation. While allowing mature mRNAs to be transported
out, the nucleus keeps unprocessed or partially processed transcripts, as well
as processed side products like removed introns, inside the nucleus. To move
across the nucleus envelope through the nuclear pore complexes, mature
mRNAs are packaged into large ribonucleoprotein (RNP) complexes. Once in
the cytoplasm, many mRNA species can be used to start synthesizing proteins
right away. As the cytoplasm is a crowded place, they may randomly drift in
the cytoplasm while translating. Some translations, however, take place at
highly localized sites. For example, in neurons some mRNAs are required
to be transported to distal dendritic regions for translation. Local protein
translation at such target sites has been known to have important biological
functions, such as synaptic plasticity that underlies learning and memory [9].
In order to transfer mRNAs to these special locations, the mRNAs bind to
special proteins to form mRNA-​protein complexes, which are then attached
to protein motors to move along cytoskeletal tracks.

3.3.6 Stability and Decay


Steady-​state mRNA concentrations, which are the detection target of
transcriptomic analysis such as RNA-​seq, are determined by not only rates of
mRNA production, but also their decay. In general, prokaryotic mRNAs are
RNA: The Transcribed Sequence 43

unstable and quickly degraded by endoribonucleases and exoribonucleases


after transcription. As a result, most of them are short-​lived and the average
prokaryotic mRNA half-​life, that is, the amount of time to have half of the
mRNAs degraded, is under 10 minutes [10]. This high turnover rate allows
prokaryotic cells to respond quickly to environmental changes by altering
transcription. In comparison, eukaryotic mRNAs are in general more stable
and have a longer average half-​life of 7–​10 hours [11, 12]. As a general rule,
mRNAs for regulatory or inducible proteins, such as transcription factors
or stress response proteins, tend to have shorter half-​lives (e.g., less than 30
minutes), while those for housekeeping proteins, such as those of metab-
olism and cellular structure, have long half-​lives (e.g., days). The stability
and half-​lives of mRNAs are also regulatable based on developmental stage
or environmental factors. For example, the stability and half-​lives of mRNAs
of muscle-​specific transcription factors, such as myogenin and myoD, are the
highest during muscle differentiation, but quickly decline once the differen-
tiation is completed [13].
The regulation of eukaryotic mRNA degradation is not well understood,
but has been known to involve interactions between some sequence elem-
ents on mRNAs and protein as well as small RNA factors. One example of the
mRNA stability regulatory sequences is the AU-​rich element, a region on the 3’
untranslated portion of many short-​lived mRNAs that, as the name suggests, is
rich in adenines and uridines. Many protein factors interact with this element
to modulate mRNA turnover, such as the AU-​rich binding factor 1 (or AUF1).
Small RNAs, including miRNA, siRNA, and piRNA, are also important
regulators of mRNA stability and degradation (see Section 3.4.4 for details).
P-​bodies (for processing bodies), granular structures in eukaryotic cells, are the
focal point of mRNA turnover mediated by protein and small RNA factors [14].
Most eukaryotic mRNA decay starts with deadenylation at the 3’ end, that
is, removal of the poly-​A tail by deadenylases. The deadenylation then leads
to mRNA degradation through two alternative mechanisms. One mechanism
is through decapping of mRNA at the 5’ end, which leaves the mRNA vul-
nerable to degradation by 5’→3’ exoribonuclease. The other mechanism is
direct 3’→5’ decay from the tail end, which is carried out by a multi-​protein
complex called the exosome complex. Besides these major deadenylation-​
dependent mRNA decay pathways, there are also other pathways that do not
rely on deadenylation, such as those dependent on 3’-​uridylation which adds
a uridine-​rich tail to the 3’ end [15].

3.3.7 Major Steps of mRNA Transcript Level Regulation


As indicated above, the regulation of both prokaryotic and eukaryotic
transcription is mostly applied at the initiation step, and this regulation is
heavily dependent on specific protein-​DNA interactions. In the prokaryotic
system, besides promoter strength, the regulation of transcriptional initiation
is provided by protein factors including repressors and activators, both of
44 Next-Generation Sequencing Data Analysis

DNA

1 - Transcriptional control

Pre-mRNA

2 – RNA processing control

mRNA
Nucleus

Cytoplasm 3 – mRNA transport & localization control

mRNA

4 – Translational control

Protein

5 – mRNA stability & degradation control

Inactive mRNA/
Degraded mRNA
FIGURE 3.3
The regulation of eukaryotic gene expression at multiple levels.

which bind to specific promoter sequences. In the eukaryotic system, specific


transcription factors that bind to specific sequences in promoters and/​
or enhancers offer most of the regulation. In addition, prior to the engagement
of transcription initiation complex, gene sequence access is regulated
through histone modification and chromatin remodeling. Since the generation
of mRNA in the eukaryotic system is a multi-​ step process, regulatory
mechanisms are also applied at subsequent steps (Figure 3.3). During mRNA
maturation, regulation of exon and intron splicing leads to generation of alter-
native splicing variants. Trafficking of mRNAs to localized cellular domains
provides additional regulatory mechanism for some genes [16]. Equally
important in determining steady-​state mRNA levels, mRNA decay is another
important but less studied step upon which regulation is also exerted.

3.4 RNA Is More Than a Messenger


Despite its apparent indispensability, mRNAs constitute only about 5%
of total cellular RNA. Besides rRNAs and tRNAs, there are also a large
RNA: The Transcribed Sequence 45

number of non-​protein-​coding RNAs that play important roles in regulating


protein-​coding genes or carry out essential cellular functions. These non-​
coding RNAs include microRNAs (or miRNAs), PIWI-​interacting RNAs
(piRNAs), small interfering RNA (siRNAs), snRNAs, small nucleolar RNAs
(snoRNAs), long-​non-coding RNAs (lncRNAs), and RNAs that function as
catalysts (ribozymes). Some of these non-​coding RNAs have been exten-
sively studied, such as the ribozymes, the discovery of which won the 1989
Nobel Prize in Chemistry and has led to the “RNA world” hypothesis. Based
on this hypothesis, early life forms were solely based on RNA, whereas DNA
and protein evolved later. The rRNAs, tRNAs, and ribozymes are thought
by this hypothesis to be evolutionary remnants of the original RNA world
[17]. The functional importance and diversity of other non-​coding RNAs,
such as the many forms of small RNAs and lncRNAs, are still in the pro-
cess of being fully appreciated because they were discovered more recently.
However, because of their wide presence and importance, the 2006 Nobel
Prize in Physiology or Medicine was awarded to the discovery of RNA inter-
ference (RNAi) by small RNAs. Due to the diverse and important roles that
non-​coding RNAs play in cells, RNA is not treated as simply a messenger
anymore.

3.4.1 Ribozyme
Similar to proteins, RNAs can form complicated three-​ dimensional
structures, and some RNA molecules carry out catalytic functions. These
catalytic RNAs are called ribozymes. A classic example of ribozyme is one
type of intron called group I intron, which splices itself out of the pre-​mRNA
that contains it. This self-​splicing process, involving two transesterification
steps, is not catalyzed by any protein. Group I intron is about 400 nucleotides
in length and mostly found in organelles, bacteria, and the nucleus of lower
eukaryotes. When a precursor RNA that contains group I intron is incubated
in a test tube, the intron splices itself out of the precursor RNA autonomously.
Despite variations in their internal sequences, all group I introns share a char-
acteristic spatial structure, which provides active sites for catalyzing the two
steps. Another example of ribozyme is the 23S rRNA contained in the large
subunit of the prokaryotic ribosome. This rRNA catalyzes the peptide bond
formation between an incoming amino acid and the existing peptide chain.
Although the large subunit contains over 30 proteins, rRNA is the catalytic
component while the proteins only provide structural support and stabiliza-
tion [18].
Also similar to protein catalysts, the dynamics of the reactions catalyzed
by ribozymes follows the same characteristics as those of protein enzyme-​
catalyzed reactions, which are usually described by the Michaelis–​Menten
equation. Further similarities of ribozymes to protein enzymes include that
ribozyme activity can also be regulated by ligands, usually small molecules,
the binding of which leads to structural change in the ribozyme. For instance,
46 Next-Generation Sequencing Data Analysis

a ribozyme may contain a riboswitch, which as part of the ribozyme can bind
to a ligand to turn on or off the ribozyme activity.

3.4.2 snRNA and snoRNA


Although group I intron can self-​splice, most pre-​mRNA introns are not of
this type and need the spliceosome for splicing. The spliceosome, even larger
than the ribosome in size, contains five snRNAs (U1, U2, U4, U5, and U6),
and a large number of proteins. The splicing process heavily depends on the
interactions between these snRNAs and pre-​mRNAs. For example, to initiate
splicing, U1 interacts with the 5’ splice donor site, and for catalysis of the
splicing process U2/​U5/​U6 comprise the active site core. High-​resolution
structural data have even shown that the spliceosome, like the ribosome, is
also a ribozyme [19].
Similarly, snoRNAs are indispensable for pre-​ rRNA processing. The
eukaryotic ribosome contains four rRNAs –​28S, 18S, 5.8S, and 5S –​with
the first three initially transcribed into a single large rRNA precursor. To
generate the three rRNAs, the precursor rRNA needs to be first chemically
modified and then cleaved. The chemical modification includes methy-
lation at over 100 nucleotides, and isomerization of uridine at another
100 sites. The snoRNAs are required in this process to identify the spe-
cific sites for modification. There are many different types of snoRNAs,
each of which can form a complementary region with the precursor rRNA
via base pairing. These duplex regions are then recognized as targets for
modification.

3.4.3 RNA for Telomere Replication


Located at the tips of a chromosome, telomeres seal the ends of chromo-
somal DNA. Without telomeres, the integrity of chromosomes would be
compromised since DNA repair enzymes would recognize the DNA termini
as break points. Inside each telomeric region is a long string of highly repeti-
tive DNA sequences. Normally shortening of telomere length occurs with
each chromosome replication, since chromosomal DNA duplication enzymes
cannot reach to the very ends of the DNA (the end replication problem). To
prevent this problem in germ cells and stem cells, an enzyme called tel-
omerase is responsible for replenishing the telomeric region. The telomerase
is a large complex comprised of an RNA component, which serves as a tem-
plate for the repeat sequence, and a catalytic protein component (reverse
transcriptase), which uses the RNA template to synthesize the repetitive
telomeric DNA sequence. The telomerase activity is usually turned off or at
very low levels in most somatic cells. Therefore, these cells can only divide
a certain number of times before reaching senescence due to the gradual
shortening of the telomere.
RNA: The Transcribed Sequence 47

3.4.4 RNAi and Small Non-​Coding RNAs


RNAi, as a cellular mechanism that uses small RNAs to silence gene expres-
sion, offers an excellent illustration of the significance of non-​coding RNAs
in the regulation of protein-​coding genes. RNAi achieves gene silencing
by suppressing mRNA translation, degrading mRNAs, or inhibiting gene
transcription [20]. As a native gene regulation mechanism in a wide-​range
of organisms, RNAi plays an essential role in organismal development and
various cellular processes. As viral RNA can activate the RNAi pathway in
host cells leading to degradation of the viral RNA, RNAi is also used by
plants and some animals to fight viral infections. Furthermore, RNAi can
also silence mobile elements in the genome, such as transposons, to maintain
genome stability. Currently large amounts of data have established the per-
vasiveness of small RNA mediated RNAi in many organisms. For example,
in the human genome, over 60% of genes are regulated by small non-​coding
RNAs [21]. Since its discovery, RNAi has been applied as a powerful research
tool to silence virtually any gene in the genome in order to decipher their
functions. Clinically, small RNAs have been tested as a strategy for gene
therapy through turning off faulty genes that underlie many genetic diseases.
RNAi is mediated by three principal groups of small non-​ coding
RNAs: miRNA, siRNA, and piRNA. All these small RNAs induce RNAi
through the same basic pathway that involves a ribonucleoprotein complex
called the RNA-​induced silencing complex (RISC). Below is a more detailed
introduction to these three groups of small RNAs and their differences.

3.4.4.1 miRNA
Mature miRNA, at around 22 nucleotides in size, induces gene silencing
through mRNA translational repression or decay. The precursor of miRNA is
usually transcribed from non-​protein-​coding genes in the genome (Figure 3.4).
The primary transcript, called pri-​miRNA, contains internal hairpin structure
and is much longer than mature miRNA. For initial processing, the pri-​miRNA
is first trimmed in the nucleus by a ribonuclease called Drosha that exists
as part of a protein complex called the microprocessor, to an intermediate
molecule called pre-​miRNA, about 70-​nucleotide in size. Alternatively, some
miRNA precursors originate from introns spliced out from protein-​coding
transcripts. These precursors, to be processed for the generation of mirtrons
(miRNAs derived from introns), bypass the microprocessor complex in the
nucleus. For further processing, the pre-​miRNA and the mirtron precursor
are exported out of the nucleus into the cytoplasm, where they are cleaved
by the endoribonuclease Dicer to form double-​stranded miRNA. The double-​
stranded miRNA is subsequently loaded into RISC. Argonaute, the core
protein component of RISC, unwinds the two miRNA strands and discard
one of them [22]. The remaining strand is used by Argonaute as the guide
sequence to identify related mRNA targets through imperfect base pairing
48 Next-Generation Sequencing Data Analysis

miRNA
Gene

Pri-miRNA

Drosha

Pre-miRNA
Exportin 5
Nucleus

Cytoplasm dsRNA

Dicer

miRNA: siRNA
miRNA* duplex
duplex Unwind

Asymmetric RISC Some


assembly miRNA

RIS C RISC
Ribosome

Target
mRNA
ORF
RISC RISC RISC
Translational repression m RNA cleavage

FIGURE 3.4
The generation and functioning of miRNA and siRNA in suppressing target mRNA activity.
Genomic regions that code for miRNAs are first transcribed into pri-​miRNAs, which are processed
into smaller pre-​miRNAs in the nucleus by Drosha. The pre-​miRNAs are then transported by
exportin 5 into the cytoplasm, where they are further reduced to miRNA:miRNA* duplex by Dicer.
While both strands of the duplex can be functional, only one strand is assembled into the RNA-​
induced silencing complex (RISC), which induces translational repression or cleavage of target
mRNAs. Long double-​stranded RNA can also be processed by Dicer to generate siRNA duplex,
which also uses RISC to break down target mRNA molecules. (Adapted by permission from
Macmillan Publishers Ltd: Nature Reviews Genetics, He L. and Hannon G.J. (2004) MicroRNAs: small
RNAs with a big role in gene regulation. Nature Reviews Genetics 5, 522–​531, ©2004.)

with seed sequence usually located in the 3’-​UTR of mRNAs. Through this
miRNA-​mRNA interaction, RISC induces silencing of target genes through
repressing translation of the mRNAs and/​or their deadenylation and degrad-
ation. Because the base pairing is imperfect, one miRNA can target multiple
RNA: The Transcribed Sequence 49

target genes’ mRNAs. Conversely, one mRNA may be targeted by multiple


miRNAs.

3.4.4.2 siRNA
While being similar in size and using basically the same system for gene
silencing (Figure 3.4), siRNA differs from miRNA in a number of aspects. On
origin, siRNA is usually exogenously introduced, such as from viral inva-
sion or artificial injection. But they can also be generated endogenously, e.g.,
from repeat-​sequence-​generated transcripts (such as those from telomeres
or transposons), or RNAs synthesized from convergent transcription (in
which both strands of a DNA sequence are transcribed from the two opposite
orientations with corresponding promoters), or other naturally occurring
sense-​antisense transcript pairs [23]. To generate mature siRNA, exogenously
introduced double-​stranded RNA, or endogenously transcribed precursor
that is transported from the nucleus to the cytoplasm, is cleaved by Dicer.
The mature siRNA is then loaded into RISC for silencing target mRNAs by
Argonaute. On target mRNA identification, siRNA differs from mRNA in that
it has perfect or nearly perfect sequence complementarity with their target.
On the mechanism of gene silencing, siRNA usually leads to endonucleolytic
cleavage, also called slicing, of the mRNAs.

3.4.4.3 piRNA
As a relatively newer class of small non-​coding RNA, piRNAs are between
23 and 31 nucleotides in length, and have functions mostly found in animal
germline tissues as a defense mechanism against transposons (or transpos-
able elements, “selfish” DNA elements that have the capability to move
around in the genome). While functioning with a similar basic RNAi mech-
anism, piRNA is different from miRNA and siRNA in two major aspects.
One is that its biogenesis does not involve Dicer, and the other is that, for
target gene silencing, it specifically interacts with PIWI proteins, a different
clade in the Argonaute protein family. The biogenesis of piRNA, independent
Dicer activity, starts from transcription of long RNAs from specific loci of the
genome called piRNA clusters. With regard to these clusters, it has been found
that while their locations in the genome do not show much change in related
species, their sequences are not conserved even in closely related species,
indicating that they are derived from invading transposable elements serving
as an adaptive genome immunity mechanism. After transcription, the long
piRNA precursor is transported out of the nucleus to the cytoplasm for pro-
cessing into mature piRNA. To induce target gene silencing, mature piRNA
is loaded into RISC that contains PIWI, which uses the piRNA sequence as
guide to silence target mRNAs by slicing. Besides this post-​transcriptional
silencing, piRNA-​ loaded mature RISC can also be transported into the
nucleus, where it finds and silences target mRNAs that are still in the process
50 Next-Generation Sequencing Data Analysis

of being transcribed. This transcription-​ level gene silencing is achieved


through interactions with other protein factors in the nucleus, and histone
modification that alters chromatin structure and gene access. The currently
best-​known function of piRNAs, through post-​transcriptional and transcrip-
tional gene silencing, is to repress transposon activity and thereby maintain
genome stability in germline cells. Transposon mobilization in the absence
of this repression can lead to DNA damage disrupting germ line develop-
ment. Non-​transposon gene targets of piRNAs have also been reported such
as those related to development.

3.4.5 Long Non-​Coding RNAs


Some non-​coding RNAs, unlike the small RNAs, are rather long with an
average length of over 200 nucleotides in their mature form. These RNAs,
called long non-​coding RNAs, have been discovered more recently and there-
fore less studied. The biogenesis of lncRNAs is somewhat similar to that of
mRNAs, as many of them are transcribed by RNA polymerase II and subject
to splicing, capping at the 5’ end, and polyadenylation at the 3’ end. Unlike
mRNAs, however, they are usually shorter with a median length of ~600
nucleotides, have fewer exons, and are generally expressed at levels lower
than those of mRNAs. Furthermore, their expression displays higher tissue
and developmental stage specificity than mRNAs, and are mostly localized
in the nucleus rather than transported to the cytoplasm.
Although they are relatively new, evidence on their importance in regu-
lating fundamental cellular functions is rapidly accumulating. They have
been known to control many steps of gene activity, including chromatin
remodeling, transcriptional regulation, mRNA processing, stability, local-
ization, and translation [24, 25]. For example, some lncRNAs, such as Xist
and HOTAIR, repress gene transcription at target genomic sites through
interacting with chromatin remodeling protein complexes [26, 27]. A class of
lncRNAs that was discovered by NGS is transcribed from enhancer regions
of protein-​ coding genes. These transcripts, called eRNAs (or enhancer
RNAs), have been shown to affect transcription of protein-​coding genes
that are regulated by the enhancers [28]. In general, lncRNAs regulate gene
activity via binding to transcription factors, repressing promoter activity, and
interacting with mRNA-​binding proteins and splicing factors. In addition,
lncRNAs can directly interact with mRNAs and thereby influence their sta-
bility and translation [29, 30]. Because of their functional importance, it is
not surprising that abnormal lncRNA expression can lead to diseases such as
cancer and aging-​related neurodegenerative disorders [31, 32].

3.4.6 Other Non-​Coding RNAs


Deep sequencing of the cellular transcriptome has led to the discovery of
other non-​coding RNAs. For example, circular RNAs, or circRNAs, exist in
many species and cell types. Unlike linear RNAs, which include all the RNA
RNA: The Transcribed Sequence 51

species introduced so far, circRNAs have their 5’ and 3’ ends joined forming
a loop structure. This structure makes them less vulnerable to attacks from
RNases and expectedly more stable. Because their widespread existence was
only unveiled with the use of RNA-​seq, the functions of most circRNAs are
still being investigated. Among currently established functions are the roles
they play in sequestering miRNA and RNA-binding proteins from their
targets, and regulating transcription, splicing, and translation events [33].
Besides the major non-​coding RNAs introduced in this chapter, there are also
other classes of non-​coding RNA species in cells that perform a remarkable
array of functions [34]. It is highly possible that new classes of non-​coding
RNAs will continue to be discovered through RNA sequencing.

3.5 The Cellular Transcriptional Landscape


Traditionally protein-​coding mRNA transcripts used to be the major targets
of transcriptional studies and as a result were often mis-​regarded as the
major component of a transcriptome. However, with the evolution of
transcriptomics technologies and as a result the discovery of the wide var-
iety of non-​coding RNAs, it has been gradually realized that protein-​coding
transcripts only constitute a minor fraction of a cell’s transcribed sequences.
Large-​scale studies on the landscape of cellular transcription, as carried out by
consortia including the FANTOM (Functional Annotation Of the Mammalian
genome) and ENCODE (Encyclopedia of DNA Elements), have revealed that
the majority of the genome is transcribed, and a large proportion of the tran-
scriptome is non-​coding RNAs [35, 36]. For example, after studying the tran-
scriptional landscape of 15 human cell lines, encompassing RNA populations
isolated from different cellular sub-​compartments including the cytosol and
the nucleus, the ENCODE consortium found that the transcription of the
genome is pervasive and 75% of genomic sequences, including those located
in gene-​poor regions, are present in transcripts. Many of the transcripts come
from intronic and intergenic regions that are not characterized currently and
therefore novel. The complex cellular RNA landscape adds further evidence
that RNA is not simply a messenger between DNA and protein.

References
1. Bedard AV, Hien EDM, Lafontaine DA. Riboswitch regulation
mechanisms: RNA, metabolites and regulatory proteins. Biochim Biophys Acta
Gene Regul Mech 2020, 1863(3):194501.
52 Next-Generation Sequencing Data Analysis

2. Ray PS, Jia J, Yao P, Majumder M, Hatzoglou M, Fox PL. A stress-​responsive


RNA switch regulates VEGFA expression. Nature 2009, 457(7231):915–​919.
3. Xu B, Zhu Y, Cao C, Chen H, Jin Q, Li G, Ma J, Yang SL, Zhao J, Zhu J et al.
Recent advances in RNA structurome. Sci China Life Sci 2022, 65(7):1285–​1324.
4. Imashimizu M, Oshima T, Lubkowska L, Kashlev M. Direct assessment of
transcription fidelity by high-​resolution RNA sequencing. Nucleic Acids Res
2013, 41(19):9090–​9104.
5. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore
SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue
transcriptomes. Nature 2008, 456(7221):470–​476.
6. Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative
splicing complexity in the human transcriptome by high-​throughput sequen-
cing. Nat Genet 2008, 40(12):1413–​1415.
7. Keegan LP, Gallo A, O’Connell MA. The many roles of an RNA editor. Nat Rev
Genet 2001, 2(11):869–​878.
8. Bratt E, Ohman M. Coordination of editing and splicing of glutamate receptor
pre-​mRNA. RNA 2003, 9(3):309–​318.
9. Pfeiffer BE, Huber KM. Current advances in local protein synthesis and syn-
aptic plasticity. J Neurosci 2006, 26(27):7147–​7150.
10. Rustad TR, Minch KJ, Brabant W, Winkler JK, Reiss DJ, Baliga NS, Sherman
DR. Global analysis of mRNA stability in Mycobacterium tuberculosis. Nucleic
Acids Res 2013, 41(1):509–​517.
11. Sharova LV, Sharov AA, Nedorezov T, Piao Y, Shaik N, Ko MS. Database
for mRNA half-​life of 19 977 genes obtained by DNA microarray analysis of
pluripotent and differentiating mouse embryonic stem cells. DNA Res 2009,
16(1):45–​58.
12. Yang E, van Nimwegen E, Zavolan M, Rajewsky N, Schroeder M, Magnasco
M, Darnell JE, Jr. Decay rates of human mRNAs: correlation with functional
characteristics and sequence attributes. Genome Res 2003, 13(8):1863–​1872.
13. Figueroa A, Cuadrado A, Fan J, Atasoy U, Muscat GE, Munoz-​Canoves P,
Gorospe M, Munoz A. Role of HuR in skeletal myogenesis through coordinate
regulation of muscle differentiation genes. Mol Cell Biol 2003, 23(14):4991–​5004.
14. Kulkarni M, Ozgur S, Stoecklin G. On track with P-​bodies. Biochem Soc Trans
2010, 38(Pt 1):242–​251.
15. Labno A, Tomecki R, Dziembowski A. Cytoplasmic RNA decay pathways –​
enzymes and mechanisms. Biochim Biophys Acta 2016, 1863(12):3125–​3147.
16. Willis DE, Twiss JL. Regulation of protein levels in subcellular domains
through mRNA transport and localized translation. Mol Cell Proteomics 2010,
9(5):952–​962.
17. Jeffares DC, Poole AM, Penny D. Relics from the RNA world. J Mol Evol 1998,
46(1):18–​36.
18. Cech TR. Structural biology. The ribosome is a ribozyme. Science 2000,
289(5481):878–​879.
19. Zhang L, Vielle A, Espinosa S, Zhao R. RNAs in the spliceosome: insight from
cryoEM structures. Wiley Interdiscip Rev RNA 2019, 10(3):e1523.
20. Wilson RC, Doudna JA. Molecular mechanisms of RNA interference. Annu
Rev Biophys 2013, 42:217–​239.
21. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are
conserved targets of microRNAs. Genome Res 2009, 19(1):92–​105.
RNA: The Transcribed Sequence 53

22. Kawamata T, Tomari Y. Making RISC. Trends Biochem Sci 2010, 35(7):368–​376.
23. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and
siRNAs. Cell 2009, 136(4):642–​655.
24. Liu X, Hao L, Li D, Zhu L, Hu S. Long non-​coding RNAs and their biological
roles in plants. Genomics Proteomics Bioinformatics 2015, 13(3):137–​147.
25. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G,
Martin D, Merkel A, Knowles DG et al. The GENCODE v7 catalog of human
long noncoding RNAs: analysis of their gene structure, evolution, and expres-
sion. Genome Res 2012, 22(9):1775–​1789.
26. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung
T, Argani P, Rinn JL et al. Long non-​coding RNA HOTAIR reprograms chro-
matin state to promote cancer metastasis. Nature 2010, 464(7291):1071–​1076.
27. Zhao J, Sun BK, Erwin JA, Song JJ, Lee JT. Polycomb proteins targeted by a short
repeat RNA to the mouse X chromosome. Science 2008, 322(5902):750–​756.
28. Li W, Notani D, Ma Q, Tanasa B, Nunez E, Chen AY, Merkurjev D, Zhang
J, Ohgi K, Song X et al. Functional roles of enhancer RNAs for oestrogen-​
dependent transcriptional activation. Nature 2013, 498(7455):516–​520.
29. Yoon JH, Abdelmohsen K, Srikantan S, Yang X, Martindale JL, De S, Huarte
M, Zhan M, Becker KG, Gorospe M. LincRNA-​p21 suppresses target mRNA
translation. Mol Cell 2012, 47(4):648–​655.
30. Gong C, Maquat LE. lncRNAs transactivate STAU1-​mediated mRNA decay
by duplexing with 3’ UTRs via Alu elements. Nature 2011, 470(7333):284–​288.
31. Yarmishyn AA, Kurochkin IV. Long noncoding RNAs: a potential novel class
of cancer biomarkers. Front Genet 2015, 6:145.
32. Ni YQ, Xu H, Liu YS. Roles of Long Non-​coding RNAs in the development
of aging-​ related neurodegenerative diseases. Front Mol Neurosci 2022,
15:844193.
33. Nisar S, Bhat AA, Singh M, Karedath T, Rizwan A, Hashem S, Bagga P, Reddy
R, Jamal F, Uddin S et al. Insights into the role of CircRNAs: biogenesis,
­characterization, functional, and clinical impact in human malignancies. Front
Cell Dev Biol 2021, 9:617281.
34. Cech TR, Steitz JA. The noncoding RNA revolution—trashing old rules to
forge new ones. Cell 2014, 157(1):77–​94.
35. Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, Oyama R,
Ravasi T, Lenhard B, Wells C et al. The transcriptional landscape of the mam-
malian genome. Science 2005, 309(5740):1559–​1563.
36. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer
A, Lagarde J, Lin W, Schlesinger F et al. Landscape of transcription in human
cells. Nature 2012, 489(7414):101–​108.
55

Part II

Introduction to
Next-​Generation
Sequencing (NGS) and
NGS Data Analysis
4 
Next-​Generation Sequencing (NGS)
Technologies: Ins and Outs

4.1 How to Sequence DNA: From First Generation to


the Next
The sequence of nucleotides in a DNA molecule can be determined in
multiple ways. Early in the 1970s, biochemists (Drs. Walter Gilbert and
Frederick Sanger) devised different methods to sequence DNA. Dr. Gilbert’s
method is based on chemical procedures that break down DNA specif-
ically at each of the four bases. Dr. Sanger’s method, on the other hand,
takes advantage of the DNA synthesis process. In this process, a new
DNA chain is synthesized base by base using sequence information on the
template (Chapter 2). The use of chemically modified nucleotides, that is
dideoxynucleotides, as irreversible DNA chain terminators in Dr. Sanger’s
method randomly stops the synthesis process at each base position, so a
series of new DNA chains of various lengths that differ by one base are
produced (Figure 4.1). Determining the lengths in single-​base resolution of
specifically broken DNA molecules in Dr. Gilbert’s method, or new DNA
chains that are randomly terminated at each of the four nucleotides in
Dr. Sanger’s method, enabled sequencing of the template DNA. Over the
years the Sanger method was further developed. The integration of automa-
tion into the process reduced human involvement and improved efficiency.
The use of fluorescently labeled terminators, instead of the radioactively
labeled terminators that were used initially, made it safer to operate and
sequence detection more robust. The improved separation of DNA chains
with the use of capillary electrophoresis, instead of slab gels, enabled high-​
confidence basecalls. All these developments had made the Sanger method
widely adopted and become the method of choice for the Human Genome
Project. Even today, this method is still widely used for single-​or low-​
throughput DNA sequencing. With the coming of NGS, this method has
become the synonym of first-​generation sequencing.
While it is robust in sequencing individual DNA fragments, the Sanger
method cannot easily achieve high-​throughput, which is the key to lowering

DOI: 10.1201/9780429329180-6 57
58 Next-Generation Sequencing Data Analysis
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 59

FIGURE 4.1
The Sanger sequencing method as originally proposed. This method involves a step for new DNA
strand synthesis using the sequencing target DNA as template, followed by sequence deduction
through resolution of the newly synthesized DNA strands. In the first step (A), the new strand
synthesis reaction mixture contains denatured DNA template, primer, DNA polymerase, and
dNTPs. Besides the dNTPs, the Sanger method is characterized by the use of dideoxynucleotides
(ddG, ddA, ddT, and ddC; the inset illustrates the structural difference between ddATP and
dATP) that are labeled with different fluorochromes. The DNA polymerase in the reaction
mixture incorporates dideoxynucleotides into the elongating DNA strand along with regular
nucleotides, but once a dideoxynucleotide is incorporated, the strand elongation terminates.
In this sequencing scheme, the ratio of these dideoxynucleotides to their regular counterparts
is controlled so that the polymerization can randomly terminate at each base position. The
end product is a population of DNA fragments with different lengths, with the length of each
fragment dependent on where the dideoxynucleotide is incorporated. These fragments are then
separated using capillary electrophoresis, in which smaller fragments migrate faster than larger
ones and as a result pass through the laser detector sooner. The fluorochrome labels they carry
enable computational deduction of the specific sequence of the original DNA. (Image adapted
from https://​comm​ons.wikime​dia.org/​w/​index.php?curid=​23264​166 by Estevezj. Used under
the Creative Commons Attribution-​Share Alike 3.0 Unported (CC BY-​SA 3.0) license (https://​
crea​tive​comm​ons.org/​licen​ses/​by-​sa/​3.0/​deed.en).)

sequencing cost, largely due to the segregation of its DNA synthesis pro-
cess and the subsequent DNA chain separation/​detection process. Its prin-
ciple of sequencing-​by-​synthesis, however, becomes the basis of many NGS
technologies, including Illumina’s reversible terminator sequencing, Pacific
Biosciences’ single-​molecule real-​time (SMRT) sequencing, ThermoFisher’s
Ion Torrent semiconductor sequencing, and the discontinued 454/​Roche’s
pyrosequencing. Different from the first-​generation method, these technolo-
gies use nucleotides with reversible terminator or other cleavable chemical
modifications, or regular unmodified nucleotides, so the new DNA strand
synthesis is not permanently terminated and therefore can be monitored as
or after each base is incorporated.
Not all NGS technologies are based on the principle of sequencing-​by-​
synthesis. For example, Oxford Nanopore sequencing and the discontinued
SOLiD sequencing from Life Technologies use nanopore sensing and
sequencing-​by-​ligation, respectively. Despite the differences in how different
NGS technologies work in principle, there is one common denominator
among them that separate them from first-​generation sequencing, which is
their massive data throughput by sequencing millions to billions of DNA
molecules simultaneously. Besides the ingenuity in the development of new
sequencing chemistries or detection schemes to be detailed next, the success
of NGS technologies in achieving extremely high throughput is also due to
modern engineering and computing feats. Advancements in microfluidics
and microfabrication make signal detection from micro-​volume of sequen-
cing reaction possible. Developments in modern optics and imaging tech-
nology enable tracking of sequencing reactions in high resolution, high
60 Next-Generation Sequencing Data Analysis

fidelity, and high speed. Some NGS platforms also rely on the decades of
progress in the semiconductor industry or more recent but rapid devel-
opment in nanopore technology (such as the Ion Torrent and Nanopore
platforms, respectively). High-​performance computing makes it possible
to process and deconvolve the torrent of signals recorded from millions of
these processes.
As different NGS technologies employ different mechanisms and imple-
mentation strategies, in the next section the specifics of some of the most
adopted NGS platforms at the time of writing (early 2022) are detailed. As
NGS technologies continue to evolve, new platforms will appear while some
current technologies become obsolete. While an overview of NGS platforms
usually becomes outdated fairly soon, the guiding principles on the analysis
of NGS data introduced in this book will remain.

4.2 Ins and Outs of Different NGS Platforms


4.2.1 Illumina Reversible Terminator Short-​Read Sequencing
4.2.1.1 Sequencing Principle
The Illumina NGS platform is by far the most popular and has generated the
largest amounts of NGS data. At the core of the Illumina sequencing tech-
nology is the employment of fluorescently labeled nucleotides with reversible
terminator [1]. As mentioned previously, this method is based on the same
basic principle of sequencing-​by-​synthesis as the Sanger method; but unlike
the Sanger method, after the incorporation of each of these specially modified
nucleotides, the terminator moiety they carry only temporarily prevents the
new DNA strand from extending. After optical detection of the incorporated
nucleotide based on its specific fluorescent label, the terminator moiety is
cleaved and thereby the new strand synthesis resumes for the next cycle of
nucleotide incorporation. For simultaneous detection of nucleotide incorpor-
ation in millions to billions of sequencing reactions, dATP, dCTP, dGTP, and
dTTP are labeled with different fluorescent labels so each nucleotide can be
detected by the different fluorescence signal they emit. The fluorescent labels
and the reversible terminator moiety are attached to the nucleotides via the
same chemical bond, so both of them can be cleaved off in a single reaction
after each nucleotide incorporation and detection cycle to prepare for incorp-
oration of the next nucleotide.

4.2.1.2 Implementation
The sequencing reaction in an Illumina NGS system takes place in a flow
cell (Figure 4.2). The fluidic channels in the flow cell, often called lanes, are
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 61

FIGURE 4.2
An Illumina sequencing flow cell. It is a special glass slide that contains fluidic channels inside
(called lanes). Sequencing libraries are loaded into the lanes for massively parallel sequencing
after template immobilization and cluster generation. In each step of the sequencing process,
DNA synthesis mixture, including DNA polymerase and modified dNTPs, is pumped into and
out of each of the lanes through their inlet and outlet ports located at the two ends.

where sequencing reactions take place and sequencing signals are collected
through scanning. The top and bottom surface of each lane is covered with
a lawn of oligonucleotide sequences that are complementary to the anchor
sequences in Illumina adapters. When sequencing libraries, prepared from
DNA through fragmentation and adapter ligation, are loaded into each of the
lanes, DNA templates in the libraries bind to these oligonucleotide sequences
and become immobilized onto the lane surface (Figure 4.3). After immobil-
ization, each template molecule is clonally amplified through an isothermal
process called “bridge amplification,” through which up to 1,000 identical
copies of the template are generated in close proximity (<1 micron in diam-
eter) forming a cluster. During sequencing, these clusters are basic detection
units, which generate enough signal intensity for basecalling.
62 Next-Generation Sequencing Data Analysis

FIGURE 4.3
Illumina sequencing process overview. (Used under license from Illumina, Inc. All Rights
Reserved.)

Under ideal conditions the simultaneous incorporation of nucleotides


to the many identical copies of sequencing templates in a single cluster is
expected to be in synchronization from step to step and therefore remain in
phase. In reality, a small percentage of templates loses sync with the majority
of molecules in the same cluster, leading to either falling behind (called
phasing), due to incomplete removal of the terminator as well as missing a
cycle, or being one or several bases ahead (pre-​phasing), due to incorporation
of nucleotides with no terminators. The existence of phasing and pre-​phasing
in a cluster leads to increased background noise and decreased basecall
quality. When more and more sequencing cycles are conducted, this problem
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 63

becomes worse. This is why platforms that are based on clonal amplification
(which also include the Ion Torrent platform to be detailed later) have
declining basecall quality scores toward the end. Eventually the decrease in
basecall quality reaches a threshold beyond which the quality scores become
simply unacceptable. The gradual loss of synchronicity is a major deter-
minant of read length for these platforms.

4.2.1.3 Error Rate, Read Length, Data Output, and Cost


The overall error rate of the Illumina sequencing method is below 1%, which
makes it one of the most accurate NGS platforms currently available. The most
common type of errors is single nucleotide substitution. On read length, all
available Illumina sequencers available at the time of writing can produce
reads of at least 150 bases. Some sequencers can generate reads of up to 250
bases (NovaSeq 6000) or 300 bases (MiSeq) in length. Besides reading from
one end of a DNA template (i.e., single-​end sequencing), Illumina sequencers
can also be read from both ends of the DNA fragment (called paired-​end
sequencing). Besides doubling the total number of sequence reads, paired-​end
sequencing also has the advantage of facilitating subsequent alignment to a ref-
erence genome (for details see next chapter) or genome assembly (Chapter 12),
thereby reducing the limitation caused by the relatively short read length.
With regard to data output, the different sequencers/​flow cells offered by
Illumina have different output levels (see Table 4.1), with the maximum of 3
Tb (Terabases) being achieved on the NovaSeq 6000 using the S4 flow cell.
Sequencing run time depends on read length and is typically in the range
of less than one day to a couple of days. On sequencing cost, depending
on sequencer and flow cell type, each Gb data costs US$4.80–​ 41.67 on
production-​scale sequencers (i.e., NovaSeq 6000 and NextSeq 2000). This cost
calculation is based on the current list price of sequencing reagents divided
by corresponding data output, so other costs, including library preparation
reagents, personnel time, sequencer depreciation, and service contract, are
not included. It should also be noted that the numbers provided here are as
of early 2022 and will change with future system updates.

4.2.1.4 Sequence Data Generation


There are three steps in the Illumina sequence data generation process.
Firstly, raw images captured after each cycle are analyzed to locate clusters
and report signal intensity, coordinates, and noise level for each cluster. This
step is conducted by the instrument control software. The output from this
step is fed into the next step of basecalling by the instrument Real-​Time
Analysis (RTA) software, which uses cluster signal intensity and noise level
to make basecalls and quality score calculation. This step also filters out low-​
quality reads. In the third step, the basecall files, or bcl files, are converted to
64 Next-Generation Sequencing Data Analysis

FASTQ files, which contain raw reads. Since multiple samples are typically
sequenced together in a multiplex fashion, demultiplexing of the sequence
data is also performed in the third step. This is typically performed using
Illumina’s bcl2fastq tool, but other tools such as IlluminaBasecallsToFastq [2]
can also be used. The demultiplexed FASTQ files in a compressed format are
what an end user typically receives from an NGS facility after the completion
of a run.

4.2.2 Pacific Biosciences Single-​Molecule Real-​Time (SMRT)


Long-​Read Sequencing
4.2.2.1 Sequencing Principle
The Pacific Biosciences’ SMRT sequencing platform is usually regarded as the
third-​generation sequencing technology, as it is sensitive enough to sequence
single DNA molecules and therefore can bypass amplification [3]. In add-
ition, this platform generates much longer reads than the Illumina NGS plat-
form, with the current read length reaching 25 kb and beyond. While it is
also based on the principle of sequencing-​by-​synthesis, different from the
Illumina method, SMRT sequencing uses nucleotides that carry distinct fluor-
escent labels linked to their end phosphate group but no terminator group.
When a nucleotide is incorporated into an elongating DNA strand, with the
cleavage of the end phosphate group (actually a pyrophosphate group as
mentioned earlier) the fluorescent label is simultaneously released, which
enables real-​time signal detection. As this process does not involve a sep-
arate step of fluorescent label releasing and detection, the sequence-​detecting
signal is recorded continuously as a movie for up to 30 hours instead of using
scanner images.

4.2.2.2 Implementation
At the core of PacBio sequencing is the SMRT cell, which carries millions of
wells, technically called zero-​mode waveguides (or ZMWs), for simultaneous
sequencing of millions of DNA templates (the current version, as of early
2022, has 8 million of ZMWs). ZMWs are essentially holes tens of nanometers
in diameter microfabricated in a metal film of 100 nm thickness, which is
in turn deposited onto a glass substrate. Because the diameter of a ZMW is
smaller than the wavelength of visible light, and the natural behavior of vis-
ible light passing through such a small opening from the glass bottom, only
the bottom 30 nm of the ZMW is illuminated. Having a detection volume
of only 20 zetpoliters (10−​21 L), this detection scheme greatly reduces back-
ground noise and enables detection of the light of different wavelengths
emitted from nucleotide incorporation into a new DNA strand.
While the SMRT platform performs single-​ molecule sequencing, the
standard library prep protocol still requires DNA samples at the µg level
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 65

to start (for lower DNA input amplification is needed). The library prep
process includes fragmentation of DNA into desired length, end repair/​A-​
tailing, and ligation of a hairpin loop adapter. This leads to the formation
of a circular structure called SMRTbell (Figure 4.4). To prepare for sequen-
cing, a SMRTbell template is annealed to a sequencing primer, and a DNA
polymerase enzyme molecule is subsequently bound to the template/​primer
structure. The template-​primer-​polymerase complex is then immobilized to
the bottom of a SMRT cell prior to sequencing.
The currently available PacBio SMRT sequencers (Sequel II/​IIe) have two
sequencing modes called continuous long reads (CLR) and circular con-
sensus sequencing (CCS). With CLR, the DNA polymerase continues to
advance along a template until it stops, thereby producing long reads in one
pass. With CCS, the DNA polymerase goes through the SMRTbell structure
multiple times and traverses both strands of the template in order to generate
a consensus read (Figure 4.4).

4.2.2.3 Error Rate, Read Length, Data Output, and Cost


The CCS mode significantly improves sequencing accuracy. For example,
with ten passes on a template the accuracy can reach 99.9%, i.e., Q30 [4]. In
PacBio terms, reads generated from the CCS mode are high-​fidelity (or HiFi)
reads. In comparison, the accuracy of CLR reads is lower at around 90%. In
general, the most common error type from SMRT sequencing is indels, with
most of them in homopolymer regions. While more accurate, CCS reads
are relatively shorter, currently in the range of 15–​25 kb. In comparison,
the length of CLR reads are typically 30–​60 kb with reported maximum
at over 200 kb. In terms of data output, CLR generates 150–​250 Gb data
in one run, while CCS produces 10–​30 Gb. Based on the current list price
of PacBio sequencing reagents, the per Gb cost of CLR reads approaches
that of Illumina high-​end sequencers, while CCS reads are still more costly
due to the multiple passes needed to generate consensus sequences (see
Table 4.1).

4.2.2.4 Sequence Data Generation


Primary data processing to generate raw basecalls is carried out on the
sequencer. This includes processing of the movie to extract sequencing
signals, basecalling from the extracted traces and pulses, and quality check of
the basecalls. To generate CLR reads, adapter sequence needs to be trimmed
from original polymerase-​generated reads. To generate CCS reads, adapter
sequence similarly needs to be removed first to generate subreads. Each
subread contains sequence that corresponds to one pass of a DNA template
from one direction. Subsequent post-​processing of the subreads collapses
multiple consecutive subreads to create a consensus sequence. All these data
generation steps take place on the sequencer.
66 Next-Generation Sequencing Data Analysis

Double-stranded DNA

Ligate adapters

Anneal primer and


bind DNA polymerase

Sequence

Subread
errors

Subreads
(passes)

Generate
consensus read CCS Read

Reference

FIGURE 4.4
PacBio sequencing library preparation and sequencing. The library prep process mostly involves
ligation of hairpin loop adapters to create the SMRTbell structure. A SMRTbell template can be
sequenced using either circular consensus sequencing (CCS, shown here) or continuous long
reads (CLR) mode. In the CCS mode, a template undergoes multiple passes to produce error-​
prone subreads in each pass followed by generation of accurate consensus reads. (Adapted by
permission from Springer Nature Customer Service Centre GmbH: Springer Nature, Nature
Biotechnology, Accurate circular consensus long-​read sequencing improves variant detection and
assembly of a human genome, Aaron M. Wenger et al., Copyright 2019.)
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 67

4.2.3 Oxford Nanopore Technologies (ONT) Long-​Read Sequencing


4.2.3.1 Sequencing Principle
Like the PacBio platform, the Oxford Nanopore platform is also a single-​
molecule sequencing platform, but the reads it produces can be even longer.
Instead of using sequencing by synthesis, it detects the order of nucleotides
off a strand of DNA (or RNA) using physical measurement when the mol-
ecule passes through a bio-​nanopore (Figure 4.5). To achieve the detection,
a voltage is first applied across both sides of the pore, which is embedded
in an electrically insulating membrane immersed in an electrolytic solution.
Because DNA/​ RNA is negative charged, the electric field drives their
movement through the pore. During this process, the nanopore serves as a
biosensor to detect the subtle change in ionic current caused by the passing of
nucleotides. Because different nucleotides result in different change patterns
in this process, the collected electrical signal is then decoded by base​calling
algorithms to reveal the underlying nucleotide sequence. It should be noted
that the detected ionic current change is emitted from 5–​9 nucleotides on a

115
dsDNA
Current (pA)

Motor protein
cis Nanopore 85
Memb rane
trans Ions TG A A A 5mCGCT A AC A A A TGA T5mCG
55
0 10 20 30 40
Time (ms)

Array of microscaffolds

Sensor chip

MinION
ASIC

Flow cell

FIGURE 4.5
Nanopore sequencing. Illustrated here is sequencing with a MinION flow cell which contains
512 channels with 4 nanopores in each channel. The electrically insulating membrane that carries
the nanopores is supported by an array of microscaffolds which is underlain by a senor chip.
There are electrodes on the sensor chip that correspond to the individual channels, and electrical
signals from the electrodes are recorded by the application-​specific integrated circuit or ASIC.
The recorded signals are then analyzed to make basecalls. (Adapted by permission from Springer
Nature Customer Service Centre GmbH: Springer Nature, Nature Biotechnology, Nanopore
sequencing technology, bioinformatics and applications, Yunhao Wang et al., Copyright 2021.)
68 Next-Generation Sequencing Data Analysis

strand of DNA/​RNA (the actual number of nucleotides detected varies with


the version of the nanopore ONT uses), not individual nucleotides [5]. To
achieve reliable detection, the speed at which the DNA/​RNA strand passes
through the pore is important, as it needs to be slowed down to a rate that
allows the electrical current change to be reliably recorded. This speed con-
trol is achieved by a motor protein situated at the orifice of the pore, and
the speed at the time of writing is usually in the range of 200–​450 bases per
second depending on the version.

4.2.3.2 Implementation
ONT currently offers three main devices at different data throughput
levels: MinION, GridION, and PromethION. MinION, at the lower end, is a
USB drive-​sized device that holds one flow cell. The GridION as a level up can
hold up to five flow cells. The flow cells used in MinION and GridION is of the
same type that contains 2,048 nanopores in 512 channels. The PromethION at
the high end has the capacity to run up to 48 flow cells simultaneously. The
flow cell used in PromethION has more capacity with 12,000 nanopores in
3,000 channels. For both the MinION/​GridION and PromethION flow cells,
in each channel only one pore can perform sequencing at a time. Besides
these flow cells, for commonly conducted, smaller-​scale sequencing, ONT
also offers a flow cell dongle called Flongle, which provides an adapter for
MinION/​GridION to allow use of smaller and lower-​cost flow cells. The
Flongle adapter has 126 channels allowing simultaneous sequencing from
126 nanopores.
ONT provides two modes of sequencing, with one for generation of long
reads (currently defined as below 100 kb) and the other for ultra-​long reads
(≥100 kb). The length of input DNA determines which mode to use. The
sample library prep process for long-​read sequencing involves fragmenta-
tion and/​or size selection (optional), end repair, A-​tailing, and ligation of
sequencing adapters. Ultra-​long sequencing library prep requires extraction
of ultra-​
high-​ molecular-​ weight DNA. While the steps involved in ultra-​
long sequencing library prep may continue to evolve, the current procedure
includes a transposition step that cleaves the template and attaches tags to the
cleaved ends simultaneously, a subsequent step to add sequencing adapters
to the tagged ends, and lastly an overnight elution of the DNA library prior
to loading into a flow cell.

4.2.3.3 Error Rate, Read Length, Data Output, and Cost


The ONT platform has the capability to sequence the entire length of DNA/​
RNA fragments brought to it. The longest read length achieved so far by
ONT is over 4 Mb. Typically for the long-read sequencing mode, the length
is 10–​100 kb, and for ultra-​long read sequencing it is 100–​300 kb. With
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 69

the newest nanopore (R10.4) available at the time of writing, the raw read
error rate is at 1% to achieve 99% (Q20) accuracy. Homopolymer error is the
most common error type in ONT sequencing. In terms of data output, the
MinION/​GridION can typically produce 10–​20 Gb data (30 Gb maximum
at the current moving speed of 250 bases/​second) from each flow cell. The
PromethION has a throughput of 50–​100 Gb (170 Gb maximum) per flow
cell. With a top loading capacity of 48 flow cells, the data output from the
PromethION can exceed that of PacBio Sequel II and Illumina NovaSeq 6000.
The cost of sequencing on the MinION/​GridION platforms is US$45–​90 per
Gb, and US$13–​40 per Gb on the PromethION. Again this calculation is based
on the list price per flow cell at the time of writing divided by the typical data
output on each platform.

4.2.3.4 Sequence Data Generation


ONT uses its MinKNOW software for device control, raw data collection,
and data processing to generate basecalls. For onboard real-​ time base-​
calling, its proprietary algorithm called Guppy is used. Unlike the Illumina
or PacBio platforms where basecalling is more mature, basecalling from
nanopore sequencing signal is still under constant improvement by ONT
and independent groups and significant progress has been made [6]. ONT
sequencers store the collected raw electrical signal in FAST5 files and can
provide sequence reads after basecalling in FASTQ files. The raw data in
FAST5 files can be used for independent basecalling, calling of DNA/​RNA
base modifications (e.g., methylation), and downstream analyses using other
software tools. As the accuracy of sequence reads is heavily dependent on the
basecall algorithm employed, reprocessing of raw data in FAST5 files over
time can lead to discovery of new information. Storing the raw data files,
however, does increase demands for storage space significantly.

4.2.4 Ion Torrent Semiconductor Sequencing


4.2.4.1 Sequencing Principle
Developed before the ONT platform, the Ion Torrent semiconductor sequen-
cing system is the first NGS platform that does not rely on chemically
modified nucleotides, fluorescence labeling, and the time-​consuming step
of image scanning, thereby achieving much faster speed, lower cost, and
smaller equipment footprint. The Ion Torrent platform sequences DNA
through detecting the H+​ ion released after each nucleotide incorporation
in the sequencing-​by-​synthesis process. When a nucleotide is incorporated
into a new DNA strand, the chemical reaction catalyzed by DNA polymerase
releases a pyrophosphate group and a H+​ (proton). The release of H+​ leads
to pH change in the vicinity of the reaction, which can be detected and used
70 Next-Generation Sequencing Data Analysis

to determine the nucleotide incorporated in the last cycle. As the change in


pH value is not nucleotide-​specific, to determine DNA sequence, each of the
substrate nucleotides (dATP, dGTP, dCTP, and dTTP) is added to the reaction
in order at different times. A detected pH change after the introduction of
a nucleotide suggests that the template strand contains its complementary
base at the last position.

4.2.4.2 Implementation
The library construction process in this technology is similar to other NGS
technologies, involving ligation of platform-​specific primers to DNA shotgun
fragments. The library fragments are then clonally amplified by emulsion
PCR onto the surface of 3-​micron diameter beads. The microbeads coated
with the amplified sequence templates are then deposited into an Ion chip.
Each Ion chip has a liquid flow chamber that allows influx and efflux of native
nucleotides (introduced one at a time), along with DNA polymerase and
buffer that are needed in the sequencing-​by-​synthesis process. For measuring
possible pH change associated with each introduction of nucleotide, there are
millions of pH microsensors that are manufactured on the chip bottom by the
employment of standard processes used in the semiconductor industry.

4.2.4.3 Error Rate, Read Length, Date Output, and Cost


The overall accuracy of the Ion Torrent platform is over 99%. The major
type of errors is indels caused by homopolymers. When the DNA template
contains a homopolymeric region, i.e., a stretch of identical nucleotides (such
as TTTTT), the signal in pH change is stronger and proportional to the number
of nucleotides contained in the homopolymer. For example, if the template
contains two Ts, the influx of dATPs will generate a pH change signal that is
about twice as strong as that generated for a single T. Accordingly the signal
for 3 Ts will be 1.5-​fold that of 2 Ts, and signal for 6 Ts will be reduced to 1.2
fold that of 5 Ts. Therefore, with the increase in the total number of the repeat
base, there is a gradual decrease in signal strength ratio that reduces the reli-
ability of calling the total number of the base correctly. It is estimated that the
current error rate for calling a 5-​base homopolymer is 3.5%.
There are currently (as of early 2022) three sequencing systems in the Ion
Torrent family: GeneStudio S5, Genexus, and PGM Dx. Of these systems, the
GeneStudio S5 can produce reads up to 600 bases in length and total output
of up to 25 Gb per chip. The Ion PGM Dx system on the other end of the spec-
trum generates 200 base reads at a total output of up to 1 Gb. The Genexus
system is more than just a sequencer, as it provides an integrated workflow
starting from nucleic acid extraction, library prep, template preparation, to
sequencing and reporting. Genexus uses its GX5 chip for sequencing, and
the cost listed on Table 4.1 is for the sequencing step only. GeneStudio S5
newgenrtpdf
Next-Generation Sequencing (NGS) Technologies: Ins and Outs
TABLE 4.1
Comparison of Current NGS Platforms
Data Output Cost per Common
Platform Read length Run time per Flow Cell Gb (US$) Error Type Error Rate

Illumina Reversible Terminator Short-​Read Sequencing


NovaSeq 6000 Up to 250 bases from each end ~13–​44 hours Up to 3,000 Gb $5–​30 Single nucleotide <1%
substitution
NextSeq 2000 Up to 150 bases from each end ~11–​48 hours Up to 360 Gb $8–​42 See above See above
MiSeq Up to 300 bases from each end ~4–​55 hours Up to 15 Gb $114–​259 See above See above
Pacific Biosciences Single Molecule Real-​Time (SMRT) Long-​Read Sequencing
PacBio Sequel II/​IIe CLR: typically 30–​60 kb, Up to 30 CLR: typically 150–​ CLR: US$5–​ Indels (mostly at CLR: 8–​13%
maximum >200 kb; hours 250 Gb; 8 per Gb; homopolymers) CCS: <1%
CCS: 15–​25 kb, maximum CCS: 10–​30 Gb CCS: US$38–​
>25 kb 113 per Gb
Oxford Nanopore Technologies (ONT) Sequencing
MinION/​ GridION 20 bases –​4.2 Mb, typically Up to 72 Typically 10–​20 Gb (30 US$45–​90 Long <1%
20–​200 bp for short reads, hours Gb max) per Gb homopolymers
50–​100 kb for long reads, over
100 kb for ultra-​long reads
PromethION See above See above Typically 50–​100 Gb US$13–​40 See above See above
(170 Gb max) per Gb
Ion Torrent Semiconductor Sequencing
Ion GeneStudio S5 200, 400, 600 bases ~3–​21.5 Up to 25 Gb per chip US$37–​698 Indels (mostly <1%
hours per Gb at homo-​
polymers)
Genexus Up to 400 bases ~14–​31 hours 24 Gb US$56 per See above <1%
Gb
Ion PGM Dx 200 bases 4.4 hours 0.6–​1 Gb US$941 per See above <1%
Gb

71
72 Next-Generation Sequencing Data Analysis

employs five chip types: Ion 510, 520, 530, 540, and 550, with the 550 chip
producing the most data (20–​25 Gb) and being the most cost effective and
the 510 having the least amount of data (0.3–​1 Gb) and being the least cost​
effective. The Ion 318 Dx chip used on the PGM Dx system, similar to the 510
chip in throughput, generates 600 Mb to 1Gb data and is suitable for running
molecular diagnostic tests that do not require a lot of data.

4.2.4.4 Sequence Data Generation


Sequencer operation and basecalling on the GeneStudio S5 and Ion PGM
Dx are provided by the Torrent Suite Software. The Genexus system has
its own software for managing the workflow and generating basecalls. The
basecalling process used by the software is similar. Raw voltage measure-
ment signal from pH change is first saved in DAT files for each cycle. After
a run all DAT files are condensed into a single WELLS file, which is then
used as input to generate basecalls. The WELLS file can be saved and used
for reanalysis. Basecalls are reported in an unmapped BAM file. Besides gen-
erating raw sequence reads, the software also provides additional analytic
functions, including read trimming and filtering, read mapping to a reference
genome, and other tertiary analyses through the use of plugins.

4.3 A Typical NGS Workflow


Despite the differences in how different NGS technologies work in prin-
ciple, the overall NGS workflow is similar. Sequencing genomic DNA, or
RNA transcripts, using these NGS technologies all involves multiple steps
(Figure 4.6). The early steps in this process are to construct sequencing
libraries from DNA or RNA molecules extracted from biological samples of
interest. As they are usually too large to be directly handled by most NGS
technologies, especially those that produce short reads, the extracted DNA
or RNA molecules often need to be broken into smaller fragments first. This
fragmentation can be achieved with different techniques, including enzym-
atic treatment, acoustic shearing, sonication, or chemical shear (typically for
RNA). The fragmentation step is usually followed by a size selection step
to collect fragments in a certain target range. If ultra-​long reads need to be
obtained from the long-​read platforms, a special extraction procedure is
required to retain high-​molecular-​weight DNA.
A key step in the sequencing library construction process is the ligation
of adapters to the two ends of DNA fragments. For RNA fragments, they
are usually converted to complementary DNA (cDNA) first before adding
the adapters. The adapters are artificial sequences that contain multiple
components including universal sequencing primer sequence(s) that initiate
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 73

Sequencing Target
(DNA or RNA)

Fragmentation*

Size Selection*

End Repair/A-Tailing
Adapter Ligation

Library Enrichment*

Sequencing

Data Analysis

FIGURE 4.6
The general workflow of an NGS experiment. For library construction, only core steps shared
by the different sequencing platforms are shown. The steps marked with asterisks are not used
in some library construction protocols. Adapters ligated to sequencing targets are specific to
each platform. There are other library construction strategies or procedures, such as use of non-​
ligation or target sequence capture, that are not shown here.

sequencing reactions on each fragment, and indexing (or “barcode”) sequences


to differentiate multiple samples when they are sequenced together. While
they generally serve similar functions in different NGS platforms, the actual
standard adapter sequences are specific to each platform. It is also possible
74 Next-Generation Sequencing Data Analysis

to design custom adapters to meet special needs as long as key adapter


sequence elements essential for a platform are in place. Prior to adapter liga-
tion, the two ends of the DNA (or cDNA) fragments need to be prepared in
an end repair step. After adapter ligation, the sequencing DNA templates in
the resultant library may need to be enriched through a PCR amplification
step using common sequences in the adapters. Alternatively, the constructed
library may be sequenced PCR-​ free without enrichment on most of the
platforms.

4.4 Biases and Other Adverse Factors That May


Affect NGS Data Accuracy
Just as a certain level of erroneous basecalls is inherent to an NGS platform,
the multiple steps that lead to the generation of sequence calls are not immune
to biases. Different from errors, biases affect accurate representation of the
original DNA or RNA population leading to higher (or lower) representa-
tion of some sequences than expected. The major source of biases in NGS is
the molecular steps involved in the library construction and the sequencing
process itself. Besides biases, there are also other potential factors that may
lead to the generation of inaccurate sequencing signals. Detailed next are the
various potential biases and other adverse factors during sequencing library
construction and sequencing that may affect NGS data accuracy. It should
be noted that while it is impossible to avoid them altogether, being aware
of their existence is the first step toward minimizing their influence through
careful experimental design and data analysis, and developing more robust
analytic algorithms.

4.4.1 Biases in Library Construction


Biases in DNA fragmentation and fragment size selection. The initial step of library
construction, i.e., DNA fragmentation, is usually assumed to be a random
process and not dependent on sequence context. This has been shown to be
not the case [7]. For example, sonication and nebulization cause DNA strand
breaks after a C residue more often than expected. After DNA fragmentation,
the size selection process may also introduce bias. For example, if gel extrac-
tion is employed for this process, the use of high gel melting temperature
favors recovery of fragments with high GC content.

Ligation biases. After fragmentation and size selection, double-​stranded DNA


fragments are usually adenylated, after end repair, at the two 3’-​ends gen-
erating 3’-​dA tails that facilitate subsequent ligation of adapters that carry
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 75

5’-​
dT overhangs and thereby avoid self-​ ligation of DNA fragments or
adapters. This AT overhang-​based adapter ligation process, however, tend to
be biased against DNA fragments that start with a T [8]. The sequencing of
large RNA species, such as mRNAs or long non-​coding RNAs, is also affected
by this bias, as cDNA molecules reverse transcribed from these species are
also subjected to the same adapter ligation process. Small RNA sequencing is
not affected by this bias, as the ligation of adapters in small RNA sequencing
library preparation is carried out prior to the reverse transcription step. The
small RNA adapter ligation step, however, introduces a different type of bias,
which affects some small RNAs in a sequence-​specific manner. Sequence spe-
cificity underlies small RNA secondary and tertiary structure, which is also
affected by temperature, concentration of cations, and destabilizing organic
agents (such as DMSO) in the ligation reaction mixture. The efficiency of
small RNA adapter ligation is influenced by their secondary and tertiary
structure [9].
PCR biases. After adapter ligation, the DNA library is usually enriched by
PCR for sequencing on most of the current NGS platforms. PCR, based on the
use of DNA polymerases, is known to be biased against DNA fragments that
are extremely GC-​or AT-​rich [10]. This can lead to variation in the coverage
of different genomic regions and under-​representation of those regions that
are GC-​or AT-​rich. While optimization of PCR conditions can ameliorate
this bias to some degree especially for high-​GC regions, this bias can only
be eliminated via adoption of a PCR-​free workflow. To achieve this, Illumina
provides PCR-​free options. For single-​molecule sequencing carried out on
the PacBio SMRT and ONT platforms, PCR amplification is typically not
required unless the input amount of starting DNA/​RNA is low.

4.4.2 Biases and Other Factors in Sequencing


Like PCR, the sequencing-​by-​synthesis process carried out by most current
NGS systems is also based on the use of DNA polymerases, which introduces
similar coverage bias against genomic regions of extreme GC or AT con-
tent. As the use of DNA polymerases is at the core of these technologies, it is
difficult to eradicate this bias completely. This bias should be kept in mind
though when sequencing genomes or genomic regions of extremely high GC
or AT content (>90%). Besides this enzymatic procedure, other aspects of the
sequencing process, including equipment operation and adjustment, image
analysis, and basecalling, may also introduce biases as well as artifacts. For
example, air bubbles, crystals, dust, and lint in the buffers could obscure
existing clusters (or beads) and lead to the generation of artificial signals.
Misalignment of the scanning stage, or even unintended light reflections, can
cause significant imaging inaccuracies. Unlike some of the inherent biases
mentioned above, these artifacts can be minimized or avoided by experienced
personnel.
76 Next-Generation Sequencing Data Analysis

The sequencing signal processing and basecalling steps may also intro-
duce bias. For example, on the Illumina platform, basecalling results can
be affected by color crosstalk, spatial crosstalk, as well as phasing and pre-​
phasing [11, 12]. On MiSeq, for instance, four images are generated from
four detection channels after each cycle, which need to be overlaid to extract
signal intensities for basecalling. This procedure is complicated by three
factors: 1) signals from the four channels are not totally independent, as there
is crosstalk between A and C, and between G and T channels, due to the over-
lapping in the emission spectra of their fluorescent labels; 2) neighboring
clusters may partially overlap leading to spatial crosstalk between adja-
cent clusters; and 3) signals from a particular cycle are also dependent on
signals from the cycles before and after, due to phasing and pre-​phasing.
While the Illumina’s proprietary software is efficient at dealing with these
factors for basecalling, there are other commercial and open-​source tools
that employ different algorithms for these tasks and generate varying results
[13]. The algorithms these methods use (including the Illumina method)
make different assumptions on signal distribution, which may not strictly
represent the collected data, and therefore introduce method-​specific bias to
basecalling.

4.5 Major Applications of NGS


4.5.1 Transcriptomic Profiling (Bulk and Single-​Cell RNA-​Seq)
NGS has replaced microarray as the major means of detecting transcriptomic
profiles and changes. The transcriptomic profile of a biological sample (such
as a cell, tissue, or organ) is determined by and reflects on its developmental
stage, internal, and external functional conditions. By sequencing existing
RNA species in the transcriptome, NGS provides answers to key questions
such as what genes are active and at what activity levels. RNA-​seq at single-​
cell level interrogates cellular heterogeneity and reveals different cell types
and states in a mixed population of cells or a tissue. Transcriptomic studies
are almost always comparative studies, contrasting one tissue/​stage/​condi-
tion with another. Besides gene-​level analysis, RNA-​seq can also be used to
study different transcripts derived from the same gene through alternative
splicing. As an integral part of the transcriptome, small RNAs can be simi-
larly studied by NGS. Analysis of bulk RNA-​seq data generated from large
and small RNA species is covered in Chapters 7 and 9, respectively. Due to
its uniqueness single-cell RNA-​seq data analysis is covered in a dedicated
chapter (Chapter 8).
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 77

4.5.2 Genetic Mutation and Variation Identification


Detecting and cataloging genetic mutation or variation among individuals
in a population is a major application of NGS. Existing NGS studies have
already shown that severe diseases such as cancer and autism are associated
with novel somatic mutations. Projects such as the 1000 Genomes Project have
revealed the great amount of genetic variation in a population that accounts
for individual differences in physical traits, disease predisposition, and drug
response. Chapter 10 focuses on data analysis techniques in a research setting
on how to identify mutations and various types of variations, and test their
associations with traits or diseases. Chapter 11 focuses on the clinical applica-
tion of NGS to identify actionable variants to guide bedside decision making.

4.5.3 De Novo Genome Assembly


Sanger sequencing used to be regarded as the golden standard for de novo
genome assembly, but more and more genomes, including large com-
plex genomes, have been assembled with NGS reads alone. Technological
advancements in the NGS arena, including the gradual increases of read
length in short-​read technologies and the maturation of long-​read technolo-
gies, have contributed to this trend. The development of new algorithms
for NGS-​ based genome assembly is another force behind this progress.
Chapter 12 focuses on how to use these algorithms to assemble a new genome
from NGS reads.

4.5.4 Protein-​DNA Interaction Analysis (ChIP-​Seq)


The normal functioning of a genome depends on its interaction with a multi-
tude of proteins. Transcription factors, for example, are among some of the
best-​known DNA-​interacting proteins. Many of these proteins interact with
DNA in a sequence-​or region-​specific manner. To determine which regions
of the genome these proteins bind to, the bound regions can be first captured
by a process called chromatin immunoprecipitation (or ChIP) and then
sequenced by NGS. ChIP-​seq can be applied to study how certain conditions,
such as a developmental stage or disease, affect the binding of protein factors
to their affinity regions. ChIP-​seq data analysis is covered in Chapter 13.

4.5.5 Epigenomics and DNA Methylation Study (Methyl-​Seq)


Chemical modifications of certain nucleotides and histones provide an
additional layer of genome modulation beyond the regulatory mech-
anism embedded in the primary nucleotide sequence of the genome. These
modifications and the modulatory information they provide constitute the
epigenome. NGS-​based epigenomics studies have revealed how monozy-
gotic twins display difference in certain phenotypes, and how changes in
78 Next-Generation Sequencing Data Analysis

epigenomic profile may lead to diseases such as cancer. Cytosine methylation


is a major form of epigenomic change. Chapter 14 covers analysis of DNA
methylation sequencing data.

4.5.6 Metagenomics
To study a community of microorganisms like the microbiome in the gut or
those in a bucket of seawater, where extremely large but unknown numbers
of species are present, a brutal force approach that involves the study of all
genomes contained in such a community is metagenomics. Recently the field
of metagenomics has been greatly fueled by the development of NGS tech-
nologies. By quickly sequencing everything in a metagenome, researchers can
get a comprehensive profile of the makeup and functional state of a micro-
bial community. Compared to NGS data generated from a single genome,
the metagenomics data is much more complicated. Chapter 15 focuses on
metagenomics NGS data analysis.

References
1. Bentley DR, Balasubramanian S, Swerdlow HP, Smith GP, Milton J, Brown
CG, Hall KP, Evers DJ, Barnes CL, Bignell HR et al. Accurate whole human
genome sequencing using reversible terminator chemistry. Nature 2008,
456(7218):53–​59.
2. Picard toolkit (https://​bro​adin​stit​ute.git​hub.io/​pic​ard/​)
3. Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, Peluso P, Rank D, Baybayan P,
Bettman B et al. Real-​time DNA sequencing from single polymerase molecules.
Science 2009, 323(5910):133–​138.
4. Wenger AM, Peluso P, Rowell WJ, Chang PC, Hall RJ, Concepcion GT, Ebler J,
Fungtammasan A, Kolesnikov A, Olson ND et al. Accurate circular consensus
long-​read sequencing improves variant detection and assembly of a human
genome. Nat Biotechnol 2019, 37(10):1155–​1162.
5. Jain M, Fiddes IT, Miga KH, Olsen HE, Paten B, Akeson M. Improved data
analysis for the MinION nanopore sequencer. Nat Methods 2015, 12(4):351–​356.
6. Rang FJ, Kloosterman WP, de Ridder J. From squiggle to basepair: computa-
tional approaches for improving nanopore sequencing read accuracy. Genome
Biol 2018, 19(1):90.
7. Poptsova MS, Il’icheva IA, Nechipurenko DY, Panchenko LA, Khodikov MV,
Oparina NY, Polozov RV, Nechipurenko YD, Grokhovsky SL. Non-​random
DNA fragmentation in next-​generation sequencing. Sci Rep 2014, 4:4532.
8. Seguin-​Orlando A, Schubert M, Clary J, Stagegaard J, Alberdi MT, Prado JL,
Prieto A, Willerslev E, Orlando L. Ligation bias in Illumina next-​generation
DNA libraries: implications for sequencing ancient genomes. PLoS One 2013,
8(10):e78575.
Next-Generation Sequencing (NGS) Technologies: Ins and Outs 79

9. Hafner M, Renwick N, Brown M, Mihailovic A, Holoch D, Lin C, Pena JT,


Nusbaum JD, Morozov P, Ludwig J et al. RNA-​ligase-​dependent biases in
miRNA representation in deep-​sequenced small RNA cDNA libraries. RNA
2011, 17(9):1697–​1712.
10. Aird D, Ross MG, Chen WS, Danielsson M, Fennell T, Russ C, Jaffe DB,
Nusbaum C, Gnirke A. Analyzing and minimizing PCR amplification bias in
Illumina sequencing libraries. Genome Biol 2011, 12(2):R18.
11. Wang B, Wan L, Wang A, Li LM. An adaptive decorrelation method removes
Illumina DNA base-​ calling errors caused by crosstalk between adjacent
clusters. Sci Rep 2017, 7:41348.
12. Whiteford N, Skelly T, Curtis C, Ritchie ME, Lohr A, Zaranek AW, Abnizova
I, Brown C. Swift: primary data analysis for the Illumina Solexa sequencing
platform. Bioinformatics 2009, 25(17):2194–​2199.
13. Cacho A, Smirnova E, Huzurbazar S, Cui X. A Comparison of base-​calling
algorithms for illumina sequencing technology. Brief Bioinform 2016, 17(5):
786–​795.
5 
Early-​Stage Next-​Generation Sequencing
(NGS) Data Analysis: Common Steps

In general, NGS data analysis is divided into three stages. In the primary
analysis stage, bases are called based on deconvolution of the optical or
physicochemical signals generated in the sequencing process. Regardless of
sequencing platforms or applications, the basecall results are usually stored
in the standard FASTQ format. Each FASTQ file contains a massive number of
reads, i.e., sequence readouts of DNA fragments sampled from a sequencing
library. In the secondary analysis stage, reads in the FASTQ files are quality
checked, preprocessed, and then mapped to a reference genome. The data
quality check or control (QC) step involves examining a number of sequence
reads quality metrics. Based on data QC result, the NGS sequencing files are
preprocessed in order to filter out low-​quality reads, trim off portions of reads
that have low-​quality basecalls, and remove adapter sequences or other arti-
ficial sequences (such as PCR primers) if they exist. Subsequent mapping (or
aligning) of the preprocessed reads to a reference genome aims to determine
where in the genome the reads come from, the critical information required
for most tertiary analysis (except de novo genome assembly). The stage of
tertiary analysis is highly application-​specific and detailed in the chapters of
Part III. This chapter focuses on steps in the primary and secondary stages,
especially on reads QC, preprocessing, and mapping, which are common and
shared among most applications (Figure 5.1).

5.1 Basecalling, FASTQ File Format, and Base Quality Score


The process of basecalling in the primary stage from fluorescence images,
movies, or physicochemical measurements is carried out with platform-​
specific, proprietary algorithms. For example, Illumina uses Bustard, a statis-
tical model-​based basecaller. ONT currently employs Guppy, based on the
use of a deep learning approach called RNNs (or recurrent neural networks).
Besides these onboard basecallers that come with the sequencers, other
stand-alone basecalling algorithms have also been developed towards the
goal of further improving accuracy [1, 2]. Increasing basecalling accuracy is

DOI: 10.1201/9780429329180-7 81
82 Next-Generation Sequencing Data Analysis

Basecalling

Data QC &
Preprocessing

Reads Mapping De Novo


to a Reference Genome Genome Assembly

Transcriptomic Analysis Protein-DNA Interaction


Metagenomics Analysis
(RNA-Seq) Analysis (ChIP-Seq)

Epigenomics & DNA Mutation and Variation


Methylation Analysis Discovery (Whole
(Methyl-Seq) Genome Resequencing)

FIGURE 5.1
General overview of NGS data analysis. The steps in the dashed box are common steps conducted
in primary and secondary analysis.

especially important for the long-read sequencing platforms, with the ONT
platform serving as a good example with active basecalling algorithm devel-
opment. Multiple machine learning-​based basecallers have been developed
by ONT, including Albacore, Scappie, Flappie, and Bonito, besides Guppy. At
the time of writing, Guppy offers faster speed than the others while maintains
relatively high accuracy, while Bonito, as the latest iteration of basecallers
from ONT, uses another deep learning approach called CNN (convolutional
neural networks) to achieve even better accuracy than Guppy but at a slower
speed. Other open-​source basecalling algorithms developed by the commu-
nity include DeepNano [3], Nanocall [4], Chiron [5], and causalcall [6]. As a
result of these algorithmic development efforts, significant progress has been
made and basecalling accuracy has been significantly increased.
Most end users do not usually intervene in the basecalling process but
rather focus on analysis of the basecalling results. Regardless of the sequen-
cing platform, basecalling results are usually reported in the universally
accepted FASTQ format. In file size, a typical compressed FASTQ file is usu-
ally in the multi-​GB range and may contain millions to billions of reads. In a
nutshell, the FASTQ format is a text-​based format, containing the sequence of
each read along with the confidence score of each base. Figure 5.2 shows an
example of one such read sequence reported in the FASTQ format.
The confidence (or quality) score, as a measure of the probability of making
an erroneous basecall, is an essential component of the FASTQ format. The
Early-Stage NGS Data Analysis: Common Steps 83

@HISEQ:131:C5NWFACXX:1:1101:3848:2428 1:N:0:CGAGGCTGCTCTCTAT
CTTTTATCAGACATATTTCTTAGGTTTGAGGGGGAATGCTGGAGATTGTAATGGGTATGGAGACATATCATATAAGTAATGCTAGG
GTGAGTGGTAGGAAG
+
BB7FFFFB<F<FBFBBFBFBFFFIFFFFIIIFF<FBFFFBFIFFBFFFIFFFBFB07<BFFF7BBFFFBFFFFFF<BFBFBBBBBB
B'77B<770<BBBBB

FIGURE 5.2
The FASTQ sequence read report format. Shown here is one read generated from an NGS
experiment. A FASTQ file usually contains millions to billions of such reads, with each
containing several lines as shown here. Line 1, starting with the symbol ‘@,’ contains sequence
ID and descriptor. Line 2 is the read sequence. Line 3 (optional) starts with the ‘+​’ symbol, which
may be followed by the sequence ID and description. Line 4 lists confidence (or quality) scores
for each corresponding base in the read sequence (Line 2). For Illumina–​generated FASTQ files,
the sequence ID in Line 1 in this example basically identifies where the sequence was generated.
This information includes the equipment (“HISEQ” in the above example), sequence run ID
(“131”), flow cell ID (“C5NWFACXX”), flow cell lane (“1”), tile number within the lane (“1101”),
x/​y-​coordinates of the sequence cluster within the tile (“3848” and “2848,” respectively). The
ensuing descriptor contains information about the read number (“1” is for single read here; for
paired-​end read it can be 1 or 2), whether the read is filtered (“N” here means it is not filtered),
control number (“0”), and index (or sample barcode) sequence (“CGAGGCTGCTCTCTAT”).

NGS basecall quality score (Q-​score) is similar to the Phred score used in
Sanger sequencing and is calculated as:

Q = −10 × log 10 PErr

where PErr is the probability of making a basecall error. Based on this equation,
a 1% chance of incorrectly calling a base is equivalent to a Q-​score of 20, and
Q30 means a 1/​1000 chance of making a wrong call. Usually for a basecall
to be reliable, it has to have a Q-​score of at least 20. High-​quality calls have
Q-​scores above 30, usually up to 40. For better visualization of Q-​scores
associated with their corresponding basecalls, they are usually encoded with
ASCII characters. While there have been different encoding scheme versions
(e.g., Illumina 1.0, 1.3, and 1.5), currently the NGS field has mostly settled on
the use of the same encoding scheme used by Sanger sequencing (Figure 5.3).
In the FASTQ example shown in Figure 5.2, the first base, C, has an encoded
Q-​score of B, i.e., 33.
To come up with the PErr, a control lane or spike control is usually used to
generate a basecall score calibration table in Illumina sequencing for lookup.
A precomputed calibration table can also be used in the absence of a control
lane and spike control. Because each platform calibrates their Q-​scores differ-
ently, if they are to be compared with each other or analyzed in an integrated
fashion, their Q-​scores need to be recalibrated. To carry out the recalibration,
a subset of reads is used that maps to regions of the reference genome that
contain no SNPs, and any mismatch between the reads and the reference
84 Next-Generation Sequencing Data Analysis

ASCII Character: ! " # $ % & ' ( ) * + , - . / 0 1 2 3 4 5 6 7 8 9 :


Quality Score: 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25

; < = > ? @ A B C D E F G H I J
26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

FIGURE 5.3
Encoding of basecall quality scores with ASCII characters. ASCII stands for American Standard
Code for Information Interchange, and an ASCII code is the numerical representation of a
character in computers (e.g., the ASCII code of the letter ‘B’ is 66). In this encoding scheme,
the ASCII character codes equal to Q-​scores plus 33. Current major NGS platforms, including
Illumina (after ver. 1.8), use this encoding scheme for Q-​score representation.

sequence is considered a sequencing error. Based on the rate of mismatch at


each base position of the reads, a new calibration table is constructed, which
is then used for recalibration. Even without cross-​platform NGS data com-
parison and integration, NGS data generated on the same platform can still
be recalibrated post mapping (see next) using the same approach, which
often leads to improved basecall quality scores.

5.2 NGS Data Quality Control and Preprocessing


After NGS data generation, the first step should be data quality check. While
this step does not directly generate biological insights, it is nonetheless essen-
tial and should be carried out carefully. Doing so will avoid production of
non-​sensical or even erroneous results in later steps and unnecessary con-
sumption of computational resources and time. In this process, the following
metrics of data quality need to be examined:

1) Q-​Scores: These can be examined in different ways. On a per-​ base


basis, this can be conducted by examining quality scores across all base
positions of all reads, from the first sequenced base to the last. As a
general trend, for platforms based on sequencing-​by-​synthesis, base
positions covered at early phases of a sequencing procedure tend to
have higher Q-​values than those sequenced later in the procedure. The
Q-​scores for even the late-​phase base positions, however, should still
have a median value of at least 20. If there is a significant Q-​score drop
in the late phase, the affected base positions need to be examined closely
and low-​quality bases should be trimmed off from affected reads. In
addition, increased percentage of N calls also helps determine loss of
basecall quality (an N is called when the base​calling algorithm cannot
call any of the four bases with confidence). Another way of inspecting
Q-​scores is by plotting the average Q-​score of each read and examining
Early-Stage NGS Data Analysis: Common Steps 85

their distribution pattern. For a successful run, the majority of reads


should have average Q-​score of over 30, and only a very small per-
centage of reads have average Q-​score below 20.
2) Percentage of each base at each position: If reads are obtained from a sequen-
cing library constructed from randomly generated DNA fragments, the
chance of observing each of the four bases at each base position should
be constant. Therefore, when plotting the percentages of each base across
all base positions, the plots for A, C, G, and T should be roughly parallel
to each other, and the overall percentage shown in each plot should
reflect the overall frequency of each base in the target library. If the plots
deviate significantly from being parallel, this indicates problem(s) in
the library construction process, such as existence of over-​represented
sequences in the library (such as rRNA in an RNA-​seq library), or non-​
random fragmentation.
3) Read length distribution: For platforms that produce reads of varying
length (such as the PacBio and ONT platforms), the distribution of
read length should also be examined. In combination with the distri-
bution of Q-​scores, this determines the total amount of useful data a
run generates. In addition, with data quality and total volume being
equal, a run that produces longer reads is more advantageous in terms
of sequence alignment or assembly than one with more relatively
short reads.

Besides examining reads quality and length distribution, other QC metrics


should also be examined, such as existence of artificial sequences including
adapters and PCR primers, or duplicated sequences based on sequence iden-
tity (sequence duplication can also be checked based on reference genome
mapping result). After inspecting sequence data quality, filtering should be
performed to remove low-​quality reads. Furthermore, low-​quality basecalls,
e.g., bases at the 3’ end that have Q-​scores below 20, as well as artificial
sequence contaminants, should also be trimmed off if they exist. While some
platforms (e.g., Illumina) perform sequence filtering by default prior to
FASTQ file generation, if the distribution of sequence Q-​scores is found to
be unsatisfactory after examination, additional filtering/​trimming may need
to be performed. Execution of these preprocessing tasks is a requirement for
high-​quality downstream analysis.
The most commonly used NGS data QC software includes FastQC [7],
NGS QC Toolkit [8], and fastp [9]. These toolkits have functional modules
to examine per-​ read and per-​base Q-​scores, base frequency distribution,
read length distribution, and existence of duplicated sequences and artificial
sequences. FastQC is written in Java and has a user-​friendly interface on most
operating systems including Windows. Developed in C/​C+​+​and employing
multi-​threading for parallel processing, fastp aims to achieve fast speed for
QC, as well as other preprocessing steps including adapter trimming, quality
86 Next-Generation Sequencing Data Analysis

filtering, per-​read quality pruning, etc. More recent NGS QC tools, such as
seqQscorer [10], apply machine learning approaches in an attempt to achieve
better understanding of quality issues and automated quality control. Tools
such as FQC Dashboard [11] and MultiQC [12] serve as aggregators of QC
results from other tools (e.g., FastQC) and present them in a single report.
After data QC, to perform stand-alone preprocessing tasks such as adapter
trimming and read filtering, tools such as cutadapt [13] and Trimmomatic
[14] are often used.
Some of the QC tools mentioned above, including FastQC and fastp, can be
used for both short and long reads. There are also tools such as NanoQC (part
of NanoPack) [15] specifically designed for long-read QC. Besides NanoQC,
NanoPack has a set of utilities for trimming, filtering, summarization, visual-
ization, etc. PycoQC [16] is another tool that provides interactive QC metrics
for ONT data. For PacBio long reads, SequelTools [17] provides QC data, as
well as other tasks such as reads filtering, summarization, and visualization.

5.3 Read Mapping
After the data is cleaned up, the next step is to map, or align, the reads
to a reference genome if it is available, or conduct de novo assembly. As
shown in Figure 5.1, most NGS applications require read mapping to a
reference genome prior to conducting further analysis. The purpose of this
mapping process is to locate origins of the reads in the genome. Compared
to searching for the location(s) of a single or a small number of sequences
in a genome by tools such as BLAST, simultaneous mapping of millions
of NGS reads, sometimes very short, to a genome is not trivial. Further
challenge comes from the fact that any particular genome from which NGS
reads are derived deviates from the reference genome at many sites because
of polymorphism and mutation. As a result any algorithm built for this task
needs to accommodate such sequence deviations. To further complicate the
situation, sequencing errors are often indistinguishable from true sequence
deviations.

5.3.1 Mapping Approaches and Algorithms


The mapping of NGS reads to a reference genome is not a new task in itself. As
indicated above, before the advent of NGS, a number of sequence alignment
algorithms already existed, the best known of which is BLAST. These aligners
use hash tables and seed-​and-​extend methods to perform the computation-
ally intensive process of aligning an individual query sequence against a
sequence database (such as GenBank). To use these methods to align the
Early-Stage NGS Data Analysis: Common Steps 87

millions of NGS reads to a reference genome, however, creates a scalability


problem, as they simply cannot scale up to the data volume, and scale down
to the short length of many NGS reads (as short read carries less informa-
tion). As a result, new generations of algorithms have been devised for the
mapping of NGS reads, either through optimizing the previous methods or
introducing new approaches.
Mapping of NGS reads can be separated into three steps (Figure 5.4). The
first step is to index the reference genome sequence in computer memory
to boost subsequent searching speed. The use of hash table is one way to
achieve this, while another approach is to use suffix-​tree based techniques
(to be detailed next). The second step is the so-​called “global positioning,”
with the goal of determining possible matching location(s) in the indexed
reference genome of seed sequences extracted from a read. The third step is
extensive alignment between the entire read and location(s) matched in Step
2 to verify the alignment and in the meantime produce alignment informa-
tion such as sequence variation and their type.
Hash table-​based reference genome indexing is often used for fast lookup
of matching locations of exact subsequences (or k-​mers) from reads. A hash
table is a data structure that stores associative data, in this case, k-​mers and
their associated genomic locations. After genomic sequences are indexed
by hashing, k-​mers are then extracted from each read and used as seeds
to search the hash table to identify their possible locations in the genome
(Figures 5.4 and 5.5). Aligners based on this approach include minimap2 [18],
SOAP (Short Oligonucleotide Alignment Program) [19], MAQ (Mapping and
Assembly with Qualities) [20], Illumina’s Isaac Genome Alignment Software
[21], and Novoalign (Commercial). Among these aligners, minimap2 uses
minimizers [22] to reduce the amount of computer memory needed to store
the hash table in order to further increase speed. It should be noted that while
most aligners use hash table to index the reference genome, some aligners
(such as MAQ) create a hash table from NGS reads instead, and in such cases
k-​mers extracted from the reference genome are used to look up matches in
the reads.
In the seed-​and-​extend approach used by BLAST, the seed used is con-
secutive sequence designed to locate near-​exact matches, which is not ideal
for aligning sequences that contain variations especially indels. To increase
alignment robustness, NGS reads aligners have migrated from the use of con-
secutive exact-​match seeds to nonconsecutive (or spaced) seeds. By allowing
space between seeds, the chance of finding a match is increased. In SOAP
and Novoalign, for example, to perform alignment using spaced seeds, the
reference genome sequence is first cut into equal-​sized small fragments and
saved in a big hash table in memory. The NGS reads are then cut in a similar
fashion into subsequences, which are searched against the reference genome
(Figure 5.5a). Computationally, these aligners are memory and processor
intensive and therefore not very fast.
88 Next-Generation Sequencing Data Analysis

FIGURE 5.4
Major steps in mapping NGS reads. In the first stage, the reference genome is indexed. This is
achieved through extracting seed sequences from the reference genome (a) and subsequently
the seed sequences are indexed using suffix tree or hash table (b). In the second stage, seed
sequences are extracted from reads (c), which are then used to search the indexed reference
genome for possible matching locations (d). In the example shown, each seed extracted from
read 1 is searched to locate their potential locations in the indexed genome. Based on their
adjacency some of the locations are excluded (red X) as such locations are unlikely to span the
read. In the last stage, the adjacent seeds are chained and the gap sequences between the seeds
are inspected for mismatches (red X), based on which pre-​alignment filters determine whether to
accept the alignment between the read and the genomic region (e). In the last step, the alignment
is subject to verification to generate alignment result including sequence differences and their
locations. (From Alser, M., Rotman, J., Deshpande, D. et al. Technology dictates algorithms: recent
developments in read alignment. Genome Biol 22, 249 (2021). https://​doi.org/​10.1186/​s13​
059-​021-​02443-​7. Used under the terms of the Creative Commons Attribution 4.0 International
License, http://​crea​tive​comm​ons.org/​licen​ses/​by/​4.0/​, © 2021 Alsher et al.)
Early-Stage NGS Data Analysis: Common Steps 89

a Spaced seeds b Burrows–Wheeler


Reference genome Short read Reference genome Short read
(> 3 gigabases) (> 3 gigabases)
Chr1 ACTCCCGT ACTCTAAT Chr1 ACTCCCGT ACTCTAAT
Chr2 Chr2
Chr3 Chr3
Chr4 Chr4
Concatenate into
Extract seeds single string

Position N
Burrows–Wheeler
Position 2 transform and indexing
CTGC CGTA AACT AATG

Bowtie index
Position 1
(~2 gigabytes)
ACTG CCGT AAAC T AAT ACTC CCGT ACTC TAAT ACTCCCGT ACTCTAAT

ACTG **** AAAC **** 1 T

**** CCGT **** TAAT Six seed 2 Look up AT


ACTG **** **** TAAT pairs per 3 “suffixes” AAT

**** **** AAAC TAAT read/ 4 of read •


ACTG CCGT **** **** fragment 5 •
**** CCGT AAAC **** 6 •
ACTCCCGT ACTCTAAT

Index seed pairs Hits identify


positions in
Seed index genome where
(tens of gigabytes) Look up each pair read is found
of seeds in index
A CTG **** AAAC ****
• Hits identify positions
• in genome where

• spaced seed pair
• is found

**** CCGT **** TAAT
A CTG**** **** TAAT Confirm hits Convert each
**** CCGT AAAC **** by checking hit back to
“**** ” positions genome location

Report alignment to user


FIGURE 5.5
NGS read mapping approaches. Panel (a) shows the approach based on spaced seed indexing.
In this illustration, spaced seeds extracted from the reference genome sequence are indexed by
a hash table. Panel (b) shows the approach based on Burrows–​Wheeler transform (BWT). In this
example, the algorithm Bowtie performs mapping by looking up reads base by base, from right
to left, against the transformed and indexed genome. (Adapted by permission from Macmillan
Publishers Ltd: Nature Biotechnology, How to map billions of short reads onto genomes, C
Trapnell and SL Salzberg, copyright 2009.)

To reduce demands on computational resources, another approach is


through the use of Burrows–​Wheeler transform (or BWT). BWT is an algo-
rithm that applies reversible transformation to a block of text (genomic
nucleotide sequence in the case of NGS data) to enable lossless data compres-
sion [23]. The transformation is achieved through a text reordering process
that can be efficiently implemented using the suffix array data structure
90 Next-Generation Sequencing Data Analysis

(a)

(b)

FIGURE 5.6
How Burrows–​Wheeler transform (BWT) works for aligning NGS reads. Panel (a) shows the
BWT procedure for a short example sequence ‘acaacg.’ Panel (b) shows how to use BWT to
identify the locations of read sequences prefixed by ‘aac.’ (Adapted by permission from Springer
Nature Customer Service Centre GmbH: Springer Nature, Genome Biology, Ultrafast and
memory-​efficient alignment of short DNA sequences to the human genome, B Langmead, C
Trapnell, M Pop, and SL Salzberg, copyright 2009.)

(Figure 5.6a). After the transformation, efficient indexing of the reference


genome sequence can be achieved using an algorithm proposed by Ferragnia
and Manzini [24]. Figure 5.6(b) provides an example of how such a suffix
array is used for lookup of k-​mers in the indexed genome. The combined use
of BWT, suffix array, and FM-​index effectively reduces the amount of com-
puter memory needed to store an indexed reference genome for fast mapping.
As an example, using this approach the indexed human genome only takes
2–​3 GB of computer memory instead of over 50 GB used by the spaced-​seed
indexing approach, and the run time is cut from hours to minutes. BWT is
employed by algorithms such as BWA (Burrows–​Wheeler Alignment) [25],
Bowtie/​Bowtie 2 [26, 27], and SOAP2 [28]. Figure 5.5b shows an example
how this approach works in the case of Bowtie.
After global position(s) of seed sequences from NGS reads are located in the
reference genome based on the use of either suffix tree or hash table, adjacent
seeds are chained and the gaps between them are evaluated for mismatches.
If this filtering process is passed and the read and the genomic region are
matched, pairwise alignment is then used to verify the alignment and gen-
erate alignment results in SAM or BAM formats (see next) (Figure 5.4). This
pairwise alignment procedure can be performed using different techniques,
Early-Stage NGS Data Analysis: Common Steps 91

among which Smith–​ Waterman, Hamming Distance, and Needleman–​


Wunsch algorithms are often used. The Smith–​Waterman algorithm is used
in commonly used tools such as STAR [29]. Smith–​Waterman is a local
alignment procedure based on dynamic programming (DP) [30]. DP was first
introduced by Needleman and Wunsch in 1970 to alignment of DNA and pro-
tein sequences towards the goal of producing an alignment for two sequences
that achieves maximum alignment score. The Needleman–​Wunsch algorithm
itself, however, is used for global alignment [31]. Alignment methods such as
Novoalign use Needleman–​Wunsch. Hamming Distance [32] is a non-​DP-​
based measure of dissimilarity between two sequences, i.e., the number of
locations at which nucleotides are different. The HD-​based method is used
by RMAP [33], Bowtie [34], and mrsFAST [35].

5.3.2 Selection of Mapping Algorithms and Reference


Genome Sequences
Table 5.1 lists some of the often-​used mapping algorithms. When selecting
aligners, factors including accuracy, speed, and computer memory require-
ment need to be considered. As these factors are usually conflicting, some
aligners put more emphasis on accuracy while others stress speed and
memory efficiency. If speed and memory efficiency are more important,
Bowtie2 is recommended. If higher accuracy is preferred, hash table-​based
tools such as Novoalign, Stampy [36], and SHRiMP2 [37] are often used. BWA
strikes a balance between speed and sensitivity. Most of these aligners are
initially developed to map very short reads, such as those of 35 nucleotides
from early Illumina sequencers. With the gradual increase in read length,
these aligners have been adapted accordingly. For instance, BWA-​MEM is an
adaptation of the original BWA algorithm for aligning longer Illumina short
reads [38].
For aligning much longer reads such as those from the PacBio and ONT
platforms, aligners designed to handle long sequences, such as minimap2,
GraphMap [39], BLASR [40], LAST [41], NGMLR [42], or Winnowmap/​
Winnowmap2 [43, 44] should be used. In general, mapping of long reads
follows a similar seed-​ chain-​
extend approach as used for short-read
aligners. For effective mapping, long reads are typically broken into shorter
subsequences that are then used as seeds for finding exact matches in the
reference genome. One of the challenges for long read mapping, however,
is the significant number of short seeds extracted from each long read. To
counter this problem and achieve fast mapping speed, a new approach based
on the use of a minimum set of representative seeds is often used. This min-
imizer approach provides a quick method for sampling and summarization
of k-​mers in a long sequence, based on which the similarity of two long
sequences can be assessed more readily. Based on this approach, if two long
sequences share identical subsequences of sufficient length, the same k-​mer
(or minimizer) will be selected for the two subsequences [22, 45]. By adopting
92 Next-Generation Sequencing Data Analysis

TABLE 5.1
Commonly Used Alignment Methods
Name Description Reference

Minimap2 A general-purpose alignment tool for both long and short reads. [18]
Uses hash table for reference genome indexing. Achieves fast
speed through use of minimizers. Splice-​aware and can be used
for long RNA-​seq reads
BWA-​MEM2 The often-​used algorithm in the BWA package designed for [48]
short reads. Employs suffix array lookup of seed sequences and
Smith–​Waterman-​based extended alignment
Bowtie2 A short-read aligner based on the use of BWT and FM [27]
for reference genome indexing, and Smith–​Waterman or
Needleman–​Wunsch for local or global alignment
SOAP2 Uses BWT compression to index the reference genome to [28]
achieve high speed for short-read alignment
Stampy A short-read aligner that has high sensitivity in mapping [36]
reads that contain variation(s) or diverge from the reference
sequence. Uses fast hashing to build reference index and a
statistical model for alignment
NGMLR A mapper designed for PacBio and ONT long reads. Splits long [42]
reads to shorter anchor sequences for lookup using hashing
and then deploys Smith–​Waterman for final alignment
GraphMap A long-read aligner that uses spaced seeds for hashing-​based [39]
index construction and lookup, and then performs graph-​based
mapping and progressive refinement to achieve alignment of
long but error-​prone reads
LAST Implements the standard seed-​and-​extend approach but with [41]
the use of adaptive seeds instead of fixed-​length seeds

minimizers, localizing subsequences shared in two long sequences becomes


more computationally tractable. To use minimap2 as an example, the step of
indexing a reference genome is achieved through indexing minimizers from
a reference genome in a hash table. To map long reads, minimizers from the
query reads are used as seeds to find exact match(es) in the reference genome.
Co-​localized seeds are then linked together as chains for the extension step.
Besides mapping algorithms, selection of reference genome sequences,
when multiple reference genome sequences are available, also affects mapping
result. By the design of most current mappers, reads that are more similar
to the selected reference sequence align better than those that deviate more
from the reference. If the deviation is sufficiently large, it might be discarded
as a mismatch. As a result, the use of different reference genome sequences
can introduce a “reference bias.” The use of any one particular reference
genome invariably introduces this bias, as a single reference genome simply
cannot accommodate sequence variations and polymorphisms that are nat-
urally present in a population or species. This bias should be kept in mind,
Early-Stage NGS Data Analysis: Common Steps 93

especially when the genetic background of the source organism is different


from the reference genome. In this situation, comparison of mapping results
from the use of different references can help select a reference that is more
appropriate. More recent mapping strategies to counter the reference bias
include use of a “major-​allele” reference genome, in which the most common
allele is used for each variant site [46]; multiple reference genomes, which is
used by the Reference Flow method [47]; or graph-​based reference genome
(see Chapter 16, Section 16.6).

5.3.3 SAM/​BAM as the Standard Mapping File Format


Mapping results generated from the various algorithms are usually stored in
the SAM or BAM file format. SAM, standing for Sequence Alignment/​Map,
has a tab-​delimited text format. It is human readable and easy to examine
but relatively slow to parse. BAM, being the compressed binary version of
SAM, is smaller in size and faster to parse. Due to their wide use, SAM/​
BAM have become the de facto standard for storing read mapping results.
The basic structure of a SAM/​BAM file is straightforward, containing a
header section (optional) and an alignment section. The header section, if it
exists, provides generic information about the SAM/​BAM file and is placed
above the alignment section. Each line in the header section starts with the
symbol “@.” For the alignment section there are 11 mandatory fields (listed
in Table 5.2). An example of the SAM/​BAM format is presented in Figure 5.7.
In the example shown in Figure 5.7, the header section contains two lines.
The first line has the two-​letter record type code HD, signifying it as the header
line, which is always the first line if present. This record has two tags: VN,
for format version, and SO, for sorting order (in this case the alignments are
sorted by coordinate). The second line is for SQ, i.e., the reference sequence
dictionary. It also has two tags SN and LN, for reference sequence name and

TABLE 5.2
Mandatory Fields in the SAM/​BAM Alignment Section
Col Field Type Description

1 QNAME String Query sequence read (or template) NAME


2 FLAG Integer bitwise FLAG
3 RNAME String Reference sequence NAME
4 POS Integer leftmost mapping POSition on the reference sequence
5 MAPQ Integer MAPping Quality
6 CIGAR String CIGAR string
7 RNEXT String Reference name of the NEXT read (For paired-​end reads)
8 PNEXT Integer Position of the NEXT read (For paired-​end reads)
9 TLEN Integer observed Template LENgth
10 SEQ String segment SEQuence
11 QUAL String ASCII of Phred-​scaled base QUALity+​33
94 Next-Generation Sequencing Data Analysis

A
Coor 12345678901234 5678901234567890123456789012345
Ref TACGATCGAAGGTA**ATGACATGCTGGCATGACCGATACCGCGACA

+r001/1 CGAAGGTACTATGA*ATG
+r002 cggAAGGTA*TATGA
+r003 TGACAT..............TACCG
-r001/2 ACCGCGACA

B
@HD VN:1.6 SO:coordinate
@SQ SN:ref LN:45
r001 99 ref 7 30 8M2I4M1D3M = 37 39 CGAAGGTACTATGAATG *
r002 0 ref 9 30 3S6M1P1I4M * 0 0 CGGAAGGTATATGA *
r003 0 ref 16 30 6M14N5M * 0 0 TGACATTACCG *
r001 147 ref 37 30 9M = 7 -39 ACCGCGACA * NM:i:1

FIGURE 5.7
The SAM/​BAM format for storing NGS reads alignment results. The alignment shown in panel
(a) is captured by the SAM format shown in panel (b). In panel (a), the reference sequence is
shown on the top with the corresponding coordinates. Among the sequences derived from it,
r001/​1 and r001/​2 are paired reads. The bases in lower cases in r002 do not match the reference
and as a result are clipped in the alignment process. The read r003 represents a spliced alignment.
In panel (b), the SAM format contains 11 mandatory fields that are explained in more detail on
Table 5.2.

reference sequence length, respectively. For the alignment section, while most
of the fields listed in Table 5.1 are self-​explanatory, some fields may not be so
clear at first glance. The FLAG field uses a simple decimal number to track
the status of 11 flags used in the mapping process, such as whether there are
multiple segments in the sequencing (like r001 in the example) or if the SEQ
is reverse complemented. To check on the status and meaning of these flags,
the decimal number needs to be converted to its binary counterpart. For the
POS field, SAM uses a 1-​based coordinate system, that is, the first base of the
reference sequence is counted as 1 (instead of 0). The MAPQ is the mapping
quality score, which is calculated similarly to the Q-​score introduced earlier
(MAPQ =​−​10 × log10(PMapErr)). The CIGAR (or Concise Idiosyncratic Gapped
Alignment Report) field describes in detail how the SEQ maps to the reference
sequence, with the marking of additional bases in the SEQ that are not
present in the reference, or missing reference bases in the SEQ. In the example
above, the CIGAR field for r001/​1 shows a value of “8M2I4M1D3M,” which
means the first eight bases matching the reference, the next two bases being
insertions, the next four matching the reference, the next one being a deletion,
and finally the last three again being matches. For more details (such as
those on the different FLAG status) and full specification of the SAM/​BAM
format, refer to the documentation from the SAM/​BAM Format Specification
Working Group. It should be noted that BAM may also be used to store
unaligned raw reads as an “off-​label” use of the format. For example, the
Early-Stage NGS Data Analysis: Common Steps 95

PacBio platform outputs raw sequencing reads in unaligned BAM format.


Besides the SAM/​BAM format, an alternative format to store aligned reads is
CRAM [49] designed by the European Bioinformatics Institute. CRAM files
are smaller than equivalent BAM files, because CRAM uses a reference-​based
compression scheme, i.e., only bases that are different from the reference
sequence are stored.

5.3.4 Mapping File Examination and Operation


After carrying out the mapping process, the mapping results reported in
SAM/​BAM files should be closely examined. Firstly, summary statistics, such
as the percentage of aligned reads, especially uniquely mapped reads, should
be generated. Currently the mapping rates are still far from 100%. Even under
ideal conditions, most aligners find unique genomic position match for 70–​
75% of sequence reads. This inability to locate the genomic origin of a sig-
nificant number of reads can be attributed to multiple factors, including the
existence of repetitive sequences in most genomes, the relatively short length
and therefore limited positioning information of most short NGS reads, algo-
rithmic limitation, sequencing error, and DNA sequence variation and poly-
morphism in a population. Mapping performance improves with increasing
read length and better-​designed algorithms from active developments in
this area.
Secondly, reads that map to multiple genomic locations, often called
multireads, usually do not contribute to subsequent analysis and therefore
should be filtered out. The ambiguity in the mapping of multireads is due
to the aforementioned sequence deviation caused by polymorphism and
mutation, sequencing error, and existence of highly similar sequences in
the genome such as those from duplicated genes. Inclusion of these reads
in downstream analysis may lead to biased or erroneous results. For most
experiments, these reads should be excluded from further analysis. As
filtering of multireads usually removes a significant number of reads, which
may lead to potential loss of information, there are some algorithms (such as
BM-​Map [50]) that are designed to reuse multireads by probabilistically allo-
cating them to competing genomic loci.
Thirdly, besides multireads, duplicate reads should also be identified and
filtered out for many experiments. In a diverse non-​enriched sequencing
library, because of the randomness of the fragmentation process the chance of
getting identical fragments is extremely low. Even with a PCR step to enrich
DNA fragments, the chance of generating duplicate reads is still very low
(usually < 5%), as the number of cycles in the PCR process is limited and the
subsequent sequencing process is a random sampling of the DNA library (to
varying depth). Existence of excessive numbers of duplicate reads therefore
suggests PCR over-​amplification. Duplicate reads can be detected based on
sequence identity, but due to sequencing error this tends to underestimate
the number of duplicate reads. It is more appropriate, therefore, to detect
96 Next-Generation Sequencing Data Analysis

FIGURE 5.8
Detection of duplicate reads after the mapping process. Depth of coverage of the reference
genomic region is shown on the top. Mapped reads, along with a set of duplicate reads that map
to the same area, are show underneath. The green and red colors denote the two DNA strands.
(Generated with CLC Genomics Workbench and used with permission from CLC Bio.)

duplicate reads after the mapping step (Figure 5.8). As technical duplicates
caused by PCR over-​amplification and true biological duplicates are indistin-
guishable, researchers should exert caution when making decisions on
whether to remove duplicate reads from further analysis. While removing
duplicate reads can lead to increased performance in subsequent analysis in
many cases (such as variant discovery), in circumstances that involve less
complex or mostly enriched sequencing targets, including those from an
extremely small genome, or those used in RNA-​seq or ChIP-​seq, removing
them can lead to loss of true biological information.
Furthermore, a variety of other steps can also be conducted to operate
SAM/​BAM files. These steps are usually provided by SAMtools and Picard,
two widely used packages for operating SAM/​BAM files. These operations
include:

• SAM and BAM interconversion. SAMtools can also convert other


alignment file formats to SAM/​BAM
• Merging of multiple BAM files into a single BAM file
• Indexing of SAM/​BAM files for fast random access
• Sorting reads alignment using various criteria, e.g., genomic coordinates,
lanes, libraries, or samples
• Additional reads alignment filtering, such as removing paired reads
that only one of the pair maps to the reference genome
Early-Stage NGS Data Analysis: Common Steps 97

ref 181 A 24 ,.$.....,,.,.,...,,,.,..^+. <<<+;<<<<<<<<<<<=<;<;7<&


ref 182 C 23 ,.....,,.,.,...,,,.,..A <<<;<<<<<<<<<3<=<<<;<<+
ref 183 A 23 ,.$....,,.,.,...,,,.,... 7<7;<;<<<<<<<<<=<;<;<<6
ref 184 G 23 ,$....,,.,.,...,,,.,...^l. <+;9*<<<<<<<<<=<<:;<<<<
ref 185 G 22 ...T,,.,.,...,,,.,.... 33;+<<7=7<<7<&<<1;<<6<
ref 186 C 22 ....,,.,.,.A.,,,.,..G. +7<;<<<<<<<&<=<<:;<<&<
ref 187 G 23 ....,,.,.,...,,,.,....^k. %38*<<;<7<<7<=<<<;<<<<<
ref 188 A 23 C..T,,.,.,...,,,.,..... ;75&<<<<<<<<<=<<<9<<:<<

FIGURE 5.9
The pileup file format as generated from SAMtools. A pileup file shows how sequenced bases in
mapped reads align with the reference sequence at each genomic coordinate. The columns are
(from left to right): chromosome (or reference name), genomic coordinate (1-​based), reference
base, total number of reads mapped to the base position, read bases, and their call qualities. In
the read bases column, dot signifies match to the reference base, comma to the complementary
strand, and ‘AGCT’ are mismatches. Additionally, the ‘$’ symbol marks the end of a read, while
‘^’ marks the start of a read and the character after the ‘^’ represents mapping quality.

• Generation of a pileup format file (Figure 5.9) to show matching (or


mismatching) bases from different reads at each genomic coordinate
(SAMtools)
• Simple visualization using a text-​based viewer for close examination of
read alignment in a small genomic region (SAMtools)

SAMtools and Picard are very versatile in handling and analyzing SAM/​
BAM files. In fact, the steps mentioned earlier, that is, generation of alignment
summary statistics and removal of multireads and duplicate reads, can be
directly conducted with these tools. For example, both SAMtools and Picard
have utilities to detect and remove duplicate reads called markdup and
markduplicates, respectively. These utilities mark reads that are mapped to
the same starting genomic locations as duplicates.
Lastly, in terms of examining mapping results, nothing can replace direct
visualization of the mapped reads in the context of the reference genome.
While a text-​based alignment viewer, such as that provided by SAMtools,
offers a simple way to examine a small genomic region, direct graphical
visualization of mapping results by overlaying mapped read sequences
against the reference genome provides a more intuitive way of examining
the data and looking for patterns. This visualization process serves mul-
tiple purposes, including additional data QC, experimental procedure valid-
ation, and mapping pattern recognition. Commonly used visualization tools
include Integrative Genomics Viewer (IGV) [51], Artemis [52], SeqMonk
[53], JBrowse [54], and Tablet [55]. The UCSC and Ensembl genome browsers
also provide visualization options by adding customized BAM tracks.
Post-​mapping data QC tools such as Qualimap 2 [56] also provide visual
summaries on key metrics including overall coverage across the reference
genome.
98 Next-Generation Sequencing Data Analysis

5.4 Tertiary Analysis
After the sequence read mapping step, subsequent analyses vary greatly
with application. For example, the workflow for RNA-​seq data analysis is
different from that for mutation and variant discovery. Therefore, it is not
possible to provide a “typical” workflow for all NGS data analyses in this
chapter beyond the common steps of data QC, preprocessing, and read
mapping. Chapters in Part III provide details on application-​specific tertiary
analytic steps and commonly used tools.

References
1. Cacho A, Smirnova E, Huzurbazar S, Cui X. A comparison of base-​ calling
algorithms for Illumina sequencing technology. Brief Bioinform 2016, 17(5):
786–​795.
2. Wick RR, Judd LM, Holt KE. Performance of neural network basecalling tools
for Oxford Nanopore sequencing. Genome Biol 2019, 20(1):129.
3. Boza V, Brejova B, Vinar T. DeepNano: deep recurrent neural networks for
base calling in MinION nanopore reads. PLoS One 2017, 12(6):e0178751.
4. David M, Dursi LJ, Yao D, Boutros PC, Simpson JT. Nanocall: an open
source basecaller for Oxford Nanopore sequencing data. Bioinformatics 2017,
33(1):49–​55.
5. Teng H, Cao MD, Hall MB, Duarte T, Wang S, Coin LJM. Chiron: translating
nanopore raw signal directly into nucleotide sequence using deep learning.
GigaScience 2018, 7(5):giy037.
6. Zeng J, Cai H, Peng H, Wang H, Zhang Y, Akutsu T. Causalcall: nanopore
basecalling using a temporal convolutional network. Front Genet 2019, 10:1332.
7. FastQC: A Quality Control Tool for High Throughput Sequence Data [Online]
(www.bio​info​rmat​ics.babra​ham.ac.uk/​proje​cts/​fas​tqc/​)
8. Patel RK, Jain M. NGS QC Toolkit: a toolkit for quality control of next gener-
ation sequencing data. PLoS One 2012, 7(2):e30619.
9. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-​fast all-​in-​one FASTQ pre-
processor. Bioinformatics 2018, 34(17):i884–​i890.
10. Albrecht S, Sprang M, Andrade-​ Navarro MA, Fontaine JF. seqQscorer:
automated quality control of next-​generation sequencing data using machine
learning. Genome Biol 2021, 22(1):75.
11. Brown J, Pirrung M, McCue LA. FQC Dashboard: integrates FastQC results
into a web-​based, interactive, and extensible FASTQ quality control tool.
Bioinformatics 2017, 33(19):3137–​3139.
12. Ewels P, Magnusson M, Lundin S, Kaller M. MultiQC: summarize analysis
results for multiple tools and samples in a single report. Bioinformatics 2016,
32(19):3047–​3048.
Early-Stage NGS Data Analysis: Common Steps 99

13. Martin M. Cutadapt removes adapter sequences from high-​ throughput


sequencing reads. EMBnet J 2011, 17(1):10–​12.
14. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina
sequence data. Bioinformatics 2014, 30(15):2114–​2120.
15. De Coster W, D’Hert S, Schultz DT, Cruts M, Van Broeckhoven C.
NanoPack: visualizing and processing long-​ read sequencing data.
Bioinformatics 2018, 34(15):2666–​2669.
16. Leger A, Leonardi T. pycoQC, interactive quality control for Oxford Nanopore
Sequencing. J Open Source Softw 2019, 4(34):1236.
17. Hufnagel DE, Hufford MB, Seetharam AS. SequelTools: a suite of tools for
working with PacBio Sequel raw sequence data. BMC Bioinformatics 2020,
21(1):429.
18. Li H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics
2018, 34(18):3094–​3100.
19. Li R, Li Y, Kristiansen K, Wang J. SOAP: short oligonucleotide alignment
program. Bioinformatics 2008, 24(5):713–​714.
20. Li H, Ruan J, Durbin R. Mapping short DNA sequencing reads and calling
variants using mapping quality scores. Genome Res 2008, 18(11):1851–​1858.
21. Raczy C, Petrovski R, Saunders CT, Chorny I, Kruglyak S, Margulies EH,
Chuang HY, Kallberg M, Kumar SA, Liao A et al. Isaac: ultra-​fast whole-​
genome secondary analysis on Illumina sequencing platforms. Bioinformatics
2013, 29(16):2041–​2043.
22. Roberts M, Hayes W, Hunt BR, Mount SM, Yorke JA. Reducing storage
requirements for biological sequence comparison. Bioinformatics 2004,
20(18):3363–​3369.
23. Burrows M, Wheeler D. A block-​sorting lossless data compression algorithm.
In: Digital SRC Research Report: 1994: Citeseer; 1994.
24. Ferragina P, Manzini G. Opportunistic data structures with applications.
In: Proceedings 41st annual symposium on foundations of computer
science: 2000: IEEE; 2000: 390–​398.
25. Li H, Durbin R. Fast and accurate long-​read alignment with Burrows–​Wheeler
transform. Bioinformatics 2010, 26(5):589–​595.
26. Langmead B, Salzberg SL. Fast gapped-​read alignment with Bowtie 2. Nat
Methods 2012, 9(4):357–​359.
27. Langmead B, Wilks C, Antonescu V, Charles R. Scaling read aligners to
hundreds of threads on general-​ purpose processors. Bioinformatics 2019,
35(3):421–​432.
28. Li R, Yu C, Li Y, Lam TW, Yiu SM, Kristiansen K, Wang J. SOAP2: an improved
ultrafast tool for short read alignment. Bioinformatics 2009, 25(15):1966–​1967.
29. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P,
Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-​ seq aligner.
Bioinformatics 2013, 29(1):15–​21.
30. Smith TF, Waterman MS. Identification of common molecular subsequences. J
Mol Biol 1981, 147(1):195–​197.
31. Needleman SB, Wunsch CD. A general method applicable to the search
for similarities in the amino acid sequence of two proteins. J Mol Biol 1970,
48(3):443–​453.
32. Hamming RW. Error detecting and error correcting codes. Bell Syst Tech J 1950,
29(2):147–​160.
100 Next-Generation Sequencing Data Analysis

33. Smith AD, Xuan Z, Zhang MQ. Using quality scores and longer reads improves
accuracy of Solexa read mapping. BMC Bioinformatics 2008, 9:128.
34. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-​efficient
alignment of short DNA sequences to the human genome. Genome Biol 2009,
10(3):R25.
35. Hach F, Hormozdiari F, Alkan C, Hormozdiari F, Birol I, Eichler EE, Sahinalp
SC. mrsFAST: a cache-​ oblivious algorithm for short-​ read mapping. Nat
Methods 2010, 7(8):576–​577.
36. Lunter G, Goodson M. Stampy: a statistical algorithm for sensitive and fast
mapping of Illumina sequence reads. Genome Res 2011, 21(6):936–​939.
37. David M, Dzamba M, Lister D, Ilie L, Brudno M. SHRiMP2: sensitive yet prac-
tical SHort Read Mapping. Bioinformatics 2011, 27(7):1011–​1012.
38. Li H. Aligning sequence reads, clone sequences and assembly contigs with
BWA-​MEM. arXiv:13033997, 2013.
39. Sovic I, Sikic M, Wilm A, Fenlon SN, Chen S, Nagarajan N. Fast and sensitive
mapping of nanopore sequencing reads with GraphMap. Nat Commun 2016,
7:11307.
40. Chaisson MJ, Tesler G. Mapping single molecule sequencing reads using basic
local alignment with successive refinement (BLASR): application and theory.
BMC Bioinformatics 2012, 13:238.
41. Kielbasa SM, Wan R, Sato K, Horton P, Frith MC. Adaptive seeds tame gen-
omic sequence comparison. Genome Res 2011, 21(3):487–​493.
42. Sedlazeck FJ, Rescheneder P, Smolka M, Fang H, Nattestad M, von Haeseler A,
Schatz MC. Accurate detection of complex structural variations using single-​
molecule sequencing. Nat Methods 2018, 15(6):461–​468.
43. Jain C, Rhie A, Zhang H, Chu C, Walenz BP, Koren S, Phillippy AM. Weighted
minimizer sampling improves long read mapping. Bioinformatics 2020,
36(Suppl_​1):i111–​i118.
44. Jain C, Rhie A, Hansen NF, Koren S, Phillippy AM. Long-​read mapping to
repetitive reference sequences using Winnowmap2. Nat Methods 2022.
45. Zheng H, Kingsford C, Marcais G. Improved design and analysis of practical
minimizers. Bioinformatics 2020, 36(Suppl_​1):i119–​i127.
46. Shukla HG, Bawa PS, Srinivasan S. hg19KIndel: ethnicity normalized human
reference genome. BMC Genomics 2019, 20(1):459.
47. Chen NC, Solomon B, Mun T, Iyer S, Langmead B. Reference flow: reducing
reference bias using multiple population genomes. Genome Biol 2021, 22(1):8.
48. Vasimuddin M, Misra S, Li H, Aluru S. Efficient architecture-​aware acceler-
ation of BWA-​MEM for multicore systems. In: 2019 IEEE International Parallel
and Distributed Processing Symposium (IPDPS): 2019: IEEE; 2019: 314–​324.
49. Hsi-​Yang Fritz M, Leinonen R, Cochrane G, Birney E. Efficient storage of
high throughput DNA sequencing data using reference-​based compression.
Genome Res 2011, 21(5):734–​740.
50. Yuan Y, Norris C, Xu Y, Tsui KW, Ji Y, Liang H. BM-​Map: an efficient software
package for accurately allocating multireads of RNA-​sequencing data. BMC
Genomics 2012, 13 Suppl 8:S9.
51. Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer
(IGV): high-​performance genomics data visualization and exploration. Brief
Bioinform 2013, 14(2):178–​192.
Early-Stage NGS Data Analysis: Common Steps 101

52. Carver T, Harris SR, Berriman M, Parkhill J, McQuillan JA. Artemis: an


integrated platform for visualization and analysis of high-​ throughput
sequence-​based experimental data. Bioinformatics 2012, 28(4):464–​469.
53. SeqMonk (www.bio​info​rmat​ics.babra​ham.ac.uk/​proje​cts/​seqm​onk/​)
54. Buels R, Yao E, Diesh CM, Hayes RD, Munoz-​Torres M, Helt G, Goodstein
DM, Elsik CG, Lewis SE, Stein L et al. JBrowse: a dynamic web platform for
genome visualization and analysis. Genome Biol 2016, 17:66.
55. Milne I, Bayer M, Cardle L, Shaw P, Stephen G, Wright F, Marshall D.
Table—next generation sequence assembly visualization. Bioinformatics 2010,
26(3):401–​402.
56. Okonechnikov K, Conesa A, Garcia-​Alcalde F. Qualimap 2: advanced multi-​
sample quality control for high-​throughput sequencing data. Bioinformatics
2016, 32(2):292–​294.
6 
Computing Needs for Next-​Generation
Sequencing (NGS) Data Management
and Analysis

The gap between our ability to pump out NGS data and our capability to
extract knowledge from these data is getting broader. To manage and pro-
cess the tsunami of NGS data for deep understanding of biological systems,
significant investment in computational infrastructure and analytical power
is needed. How to gauge computing needs and build a system to meet the
needs, however, poses serious challenges to small research groups and even
large research organizations. To meet this unprecedented challenge, the NGS
field can borrow solutions from other “big data” fields such as high-​energy
particle physics, climatology, and social media. For biologists without much
training in bioinformatics, while getting expert help is needed, having a good
understanding of the various aspects of NGS data management and analysis
is beneficial for years to come.

6.1 NGS Data Storage, Transfer, and Sharing


NGS has itself become a major producer of big data in scientific research.
With the continuous drop in sequencing cost, the speed at which NGS data is
pumped out will only pick up. This translates into a concomitant increase in
the demand for more data storage, access, and processing power. Compared
to files generated from other biological assays, such as gel pictures or even
microarray data files, NGS files are much larger. For an individual lab, a
single typical run generates data at the level of tens to thousands of gigabytes
(GB) in compressed FASTQ format. After aligning to a reference genome,
the processed files increase in size appreciably. Further analysis leads to
the generation of more and more files and propagation in data volume. To
accommodate raw and processed files from multiple runs, tens of terabytes
(TB) of storage space or more is required. Storing and archiving these files
are no trivial task. To make the situation even worse, the raw sequencing

DOI: 10.1201/9780429329180-8 103


104 Next-Generation Sequencing Data Analysis

signal intensity files in formats such as scanned images or movies are in the
scale of TB from a single run (this amount is not counted in the data volume
mentioned above). As these raw signal files accumulate, they can easily
overwhelm most data storage systems. While these raw images files can be
retained long term, newer sequencing systems process them on-​the-​fly and
delete them by default once they have been analyzed to alleviate the burden
of storing them. Oftentimes it is easier and more economical to rerun the
samples in case of data loss rather than archiving these huge raw signal files.
Due to the huge size of most NGS files, transferring them from one place
to another is non-​trivial. For a small-​sized project to transfer sequencing files
from a production server to a local storage space, download via FTP or HTTP
might be adequate if a fast network connection is available. As for network
speed, a 1 Gbps network is essential while a 10/​100 Gbps network offers
improved performance for high traffic conditions. When the network speed
is slow or the amount of data to be transferred is too large, the use of external
hard drive might be the only option. When the data reaches the lab, for fast
local file reading, writing, and processing, they need to be stored in a hard
drive array inside a dedicated workstation or server.
For a production environment, such as an NGS core facility or a large genome
center, which generates NGS data for a large number of projects, enterprise-​
level data storage system, such as DAS (Directly Attached Storage), SAN
(Storage Area Network), or NAS (Network Attached Storage), is required to
provide centralized data repositories with high reliability, access speed, and
security. To avoid accidental data loss, these data storage systems are usu-
ally backed up, mirrored, or synced to data servers distributed at separate
locations. For large-​scale collaborative projects that involve multiple sites
and petabytes to exabytes of data, the processes of data transfer and sharing
pose more challenges, which prompts the development of high-​capacity and
high-​performance platforms such as Globus.
Data sharing among collaborating groups creates additional technical
issues beyond those dealt with by individual labs. A centralized data reposi-
tory might be preferred over simple data replication at multiple sites to foster
effective collaboration and timely discussion. Along with data sharing come
also the issues of data access control and privacy for data generated from
patient-​oriented studies. In a broader sense, NGS data sharing with the
entire life science community also increases the value of a research project.
For this reason, many journals enforce a data sharing policy that requires
deposition before publication of sequence read data and processed data into
a publicly accessible database (such as the NCBI’s Sequence Read Archive
[SRA] or the European Nucleotide Archive [ENA]). To facilitate data inter-
pretation and potential meta-​analysis, relevant information about such an
experiment must also be deposited with the data. Some organizations, such
as the Functional Genomics Data Society, have developed guidelines on what
information should be deposited with the data. For example, the MINSEQE
Computing Needs for NGS Data Management and Analysis 105

(Minimum Information about a high-​throughput Nucleotide SEQuencing


Experiment) guidelines specify the following information to be provided
with the sequence read data and processed data: (1) description of the bio-
logical system, samples, and experimental variables; (2) experimental sum-
mary and sample data relationships; and (3) essential experimental and data
processing protocols. Archiving NGS data and associated information for
the community is a huge undertaking and requires sizeable investment in
maintaining and growing the requisite infrastructure and expert support.
The NCBI SRA repository was shut down in 2011 due to high costs and gov-
ernment budgetary constraints. However, because its vital importance to the
community, NIH resumed its support to SRA later that year.

6.2 Computing Power Required for NGS Data Analysis


Processing the large volume of NGS data requires a lot of computing power.
The question of how much computing power is needed is dependent on
the type of analysis to be performed. For example, de novo assembly of a
large genome requires much more computing power than resequencing
for variant discovery, or transcriptomic analysis for the identification of
differentially expressed genes. Therefore, to determine the computing
power needed for a project, a lab, or an organization, the type(s) of NGS
work to be performed need to be analyzed first. If the work will require
intensive computation, or involve development and optimization of new
algorithms and software tools, a high-​performance cluster may be needed.
On the other hand, if the work will use established workflow that does not
require highly intensive computation, a powerful workstation may suffice.
It is also advisable that the computer system be scalable to accommodate
increases in future computing needs due to unforeseeable change of future
research projects or further development of high-​throughput genomics
technologies.
For a small-​sized project, the most basic system needed for NGS data ana-
lysis can be simply a 64-​bit computer with 8 GB of RAM and two 2-​GHz
quad-​core processors. With such a computer, basic mapping to a reference
genome can be performed on obtained sequence reads. This basic setup
allows handling of one dataset at a time. For simultaneous processing of
multiple datasets or projects, high-​performance computing (HPC) systems
with more memory and CPU cores are needed. The number of cores an HPC
system needs depends on the number of simultaneous tasks to be run at one
time. For each task, the number of cores that is needed relies on the nature of
the task and the algorithm that carries it out.
106 Next-Generation Sequencing Data Analysis

Besides the number of CPU cores, the amount of memory a system has also
heavily affects its performance. Again memory needs depend on the number
and complexity of jobs to be processed, for example, read mapping to a small
genome may need only a few GB of memory while de novo assembly of a large
genome may require hundreds of GB or even TB-​level memory. The current
estimation is that for each CPU core the amount of memory needed should
not be less than 3 GB. In an earlier implementation of de novo assembly of the
human genome using the SOAPdenovo pipeline (to be detailed in Chapter 12),
a standard supercomputer with 32 cores (eight AMD quad-​core 2.3 GHz CPUs)
and 512 GB memory was used [1]. As a more recent example of the computing
power needed for de novo genome assembly, a server with 64 cores (eight
Intel Xeon X6550 8-​core 2.00 GHz CPUs) and 2 TB RAM is used by a Swedish
team [2]. For de novo assembly of small genomes such as those of microbes, a
machine that contains at least 8 CPU cores, 256 GB of RAM, and a fast data
storage system can get a job completed in a reasonable time frame. By current
estimation, an 8-​core workstation with 32 GB RAM and 10 TB storage can work
for many projects that do not conduct de novo genome assembly.
The amount of time needed to complete a job varies greatly with the com-
plexity of the job and accessible computing power. As a more concrete example,
running the deep learning-​based WGS variant calling tool DeepVariant (see
Chapter 10 for details) needs 24–​48 hours when using the minimum setting
of a 8-​core computer with 16 GB RAM, but the processing time is reduced
by more than half when using a graphics processing unit (GPU) with 4 GB
dedicated video RAM and CUDA support for parallel computing [3]. To map
an RNA-​seq dataset of 80 million 75-​bp reads to the human genome using
Bowtie on a computer equipped with 32 cores and 128 GB RAM, it took <2
hours and even less time in subsequent steps including normalization and
differential expression statistical tests [4]. In a small RNA-​seq study, with a
32-​core and 132 GB memory workstation, processing 20 multiplex barcoded
samples with a total of 160 million reads took a little over 2 hours for sample
de-​multiplexing, and about the same amount of time for read mapping to the
host genome and small RNA annotation databases [5].

6.3 Cloud Computing
As clearly demonstrated above, NGS data storage, transfer, and sharing are
no trivial tasks. And one limitation of a locally built computing system is its
scalability. With the rates at which NGS technologies advance and sequen-
cing costs drop being faster than those of development in the computer
hardware industry, the gap between NGS data generation and our ability
to handle and analyze them will only widen. To narrow this gap and speed
up NGS data processing, the NGS community has embraced a trend from
Computing Needs for NGS Data Management and Analysis 107

the long-​existing model of local computing to cloud computing. Companies


such as Amazon, Microsoft, and Google have been building mega-​scale cloud
computing clusters and data storage systems for end users to use over the
Internet. Compared to local computing, cloud computing enables access to
supercomputing and mass data storage capabilities without the need to build
and maintain a local workstation, server, or HPC cluster.
At the core of cloud computing is virtualization technology, which allows
an end user to create a virtual computer system on demand with the flexibility
of specifying the number of CPU cores, memory size, disk space, as well as
operating system that are required for a job. With this technology, multiple
virtual computer systems can be run simultaneously on the same physical
cloud server. The adoption of cloud computing for NGS data processing has
demonstrated the advantages of this “supercomputing-​on-​demand” model,
which include flexibility, scalability, and oftentimes cost savings. The flexibility
and scalability offered by cloud computing allow a researcher to conduct NGS
data analysis using supercomputing capabilities that previously only existed
in large genome centers. Cost savings are achieved as the user only needs to
pay for the time used by the user-​configured computing instance.
Another advantage of using the cloud is on data sharing among researchers
and projects. By providing a single centralized data storage, the cloud enables
different groups located in different geographical locations to have access
to the same dataset and share analytical results. Furthermore, with cloud
computing the task of bringing software tools to the “big” NGS data can be
realized more readily. In contrast to the large sizes of NGS data files, the soft-
ware and scripts designed to process them are much smaller. Therefore, it is
much easier and more efficient to download and install them to wherever the
data is stored, rather than moving or replicating the high volumes of NGS
data to where the tools are installed. By directly storing production data in
the cloud, the burden of data transfer is greatly reduced; by coupling data
and tools in the same place, optimal performance can be achieved.
While cloud computing enables users to off-​load the hassle and cost of
running and maintaining a local computing system, it does have downsides
that need to be considered. One of the practical barriers of moving to the cloud
is the speed of data transfer into and out of the cloud. It may take a week to
upload 100 GB of data to the cloud using low-​speed Internet connections.
The question of whether to run analysis in the cloud is heavily dependent
on the amount of data to be transferred and the computational complexity of
the analytical steps. As a general rule, it is only worthwhile to upload data
to the cloud for processing when the analytical task requires more than 105
CPU cycles per byte of data [6]. So for projects that deal with large amounts
of data but do not involve a lot of highly intensive computational steps, more
time may be spent on data transfer to the cloud rather than data processing.
Other potential factors include data security, cost ineffectiveness under some
circumstances, availability of analytical tools in the cloud environment, and
network downtime. While users can access their data from anywhere on the
108 Next-Generation Sequencing Data Analysis

TABLE 6.1
Current Providers of Cloud Computing That Can Be Used for NGS Data Analysis
Provider URL

Amazon Elastic Compute Cloud http://​aws.amazon.com/​ec2/​


Google Cloud https://​cloud.google.com
Microsoft Azure http://​azure.microsoft.com/​
Rackspace www.rackspace.com
IBM Cloud www.ibm.com/​cloud

Internet, the convenience also means the possibility of data security being
breached or compromised. Some heavy users may find cloud computing
not as cost effective as running a local server. While more and more tools
are becoming available in the cloud, users still need to use due diligence to
make sure that the tools they need are available. For users at places that suffer
frequent network outage, cloud computing can be problematic as all cloud-​
based operations are dependent on Internet traffic.
Despite the potential downsides, cloud computing has been proven to be
a viable approach for NGS data analysis. Table 6.1 is a list of some of the
current cloud computing providers that can be used for NGS applications.
To illustrate how cloud computing can be deployed for analyzing NGS data,
below is an example on the conduct of reads alignment using the Amazon
Elastic Compute Cloud (or EC2) Cloud. As the first step, input data files
(FASTQ files and a reference genome file) are uploaded from a local com-
puter to a “bucket” in the Amazon Simple Storage Service (S3). This cloud
storage bucket, which is also used to hold program scripts and output files,
can be created with the Amazon Web Services (AWS) Management Console, a
unified interface to access all Amazon cloud resources. To initiate alignment,
a workflow must be defined first using the Console’s “create workflow”
function. To define the workflow, the input sequence read files, the aligner
script, and the saving location for alignment output files are specified. In the
meantime, the number of Amazon EC2 instances required for the job, which
determines memory and processor allocation, is also configured. After the
configuration the job is submitted through the Management Console. When
the instances are finished, alignment output files are deposited into the pre-​
specified file location in the S3 cloud storage.

6.4 Software Needs for NGS Data Analysis


To configure a cloud instance or set up a local workstation or server, oper-
ating system and software need to be selected and installed. While some NGS
Computing Needs for NGS Data Management and Analysis 109

analysis software (such as CLC Genomics Workbench) can operate in the


Windows environment, most tools only operate in the Unix (or Linux) envir-
onment. Therefore, Unix or Linux is usually the operating system installed
on such a machine. Installing software in Unix or Linux is not as straight-
forward as in Windows, as uncompiled software source code downloaded
from a developer site needs to be compiled first before being installed to a
particular distribution of the operating system. If the reader is not familiar
with the Unix/​Linux environment and the command line interface it uses, an
introductory book or web-​based tutorial is suggested.
One approach to reducing the barrier of using tools developed for the Unix/​
Linux environment is to access them through a “bridging” system, such as
Galaxy [7], that provides a more user-​friendly interface to the command line
tools. Developed by the Nekrutenko lab at Penn State and the Taylor lab at
Johns Hopkins University, the Galaxy system provides a mechanism to deploy
these tools via the familiar web browser interface, making them accessible
to users regardless of the operating system they use. The Galaxy system is
highly extensible, with latest tools being constantly wrapped for execution
through the web interface. Besides providing a user-​friendly interface, such
a system also allows creation of data analysis workflow from different tools,
which enables fast deployment of multiple tools in tandem, achievement of
consistency and reproducibility, and sharing of analytical procedures with
other researchers. Galaxy can be accessed through a publicly available server
(e.g., usegalaxy.org), installed on a local instance, or in the cloud. With a
public server, the user does not need to maintain a local server, but the usable
storage space assigned to each account is usually limited and the computing
resource is shared with many other users. Creating a local Galaxy instance
in Unix/​Linux or Mac OS takes some effort and the user does need to pro-
vide maintenance, but the user has more control on storage space, computing
power, and selection and installation of tools from the entire collection of gen-
omics tools that are made available through the Galaxy Tool Shed (currently
has close to 10,000 tools). The Galaxy team has made it very easy to install a
local instance by offering detailed and easy-​to-​follow instructions. Galaxy is
also available in the cloud via dedicated instances in AnVIL, a cloud-​based
platform for data analysis, storage, and management built on the Google
Cloud Platform (GCP) and supported by the U.S. National Human Genome
Research Institute [8]. Besides providing a highly scalable computing envir-
onment, the Galaxy cloud instance in AnVIL also offers protection to private
datasets while facilitates collaboration.
There are also other community projects that provide alternative platforms
to facilitate user access to various NGS and other genomics analysis tools.
Bioconductor, an open-​source and open-​development software project, is
among the best known of these projects. This large-​scale project is based on R,
a programming language and software environment designed for statistical
computing and graphics. With the goal of providing tools for the analysis
110 Next-Generation Sequencing Data Analysis

and comprehension of high-​throughput genomics data, the Bioconductor


software library contains over 2,200 software packages as the time of this
writing, many of which are designed or can be used to process NGS data.
The R environment and the Bioconductor library can be installed in all major
operating systems including Windows. The Bioconductor project web portal
(www.bioco​nduc​tor.org) and the R project site (www.r-​proj​ect.org) pro-
vide detailed information and tutorials for the installation and use of these
packages. Each tool is well documented with actual use examples provided.
Identifying, installing, and maintaining suitable NGS analysis software
from an ever-​growing number of tools for a local Unix/​Linux workstation, a
local Galaxy instance, or a local Bioconductor R library are not trivial. New
software tools are developed and introduced constantly, while many existing
ones are updated from time to time. To evaluate candidate packages and iden-
tify appropriate tools for installation and use, it is better to employ multiple
test datasets, not just using those from computer simulation but also those
from real-​world biological samples. In addition, almost all tools have adjust-
able parameters, which should be set equivalently to facilitate performance
comparison. Also in terms of performance, earlier NGS software usually does
not take advantage of high-​performance parallel computing. To increase per-
formance and take full advantage of the multiple cores or nodes in an HPC
system, more recent algorithms tend to use threading or message passing
interface (MPI) to spread the work across multiple processes. Therefore,
when evaluating NGS tools, it also helps to examine if these types of parallel
processing are employed to take advantage of the power of multi-​core com-
puting architecture.

6.4.1 Parallel Computing
Parallelization, a computation term that describes splitting of a task into a
number of independent subtasks, can significantly increase the processing
speed of highly parallelizable tasks, which include many NGS data analysis
steps. For example, although millions of reads are generated from a sequen-
cing run, mapping of these reads to a reference genome is a process that is
“embarrassingly parallel,” as each read is mapped independently to the ref-
erence. As parallel computing can be efficiently carried out by GPUs since
rendering of each pixel on a computer screen is also a highly parallel process,
the integration of GPUs with CPUs in heterogeneous computing systems
can increase throughput 10-​to 100-​fold, and turn individual computers into
mini-​supercomputers. While these systems can be applied to various aspects
of NGS data analysis, many NGS analytical tools have yet to take full advan-
tage of the power of parallel computing in such systems.
Parallelization is also an important factor in determining how increase in
the number of CPU (or GPU) cores might affect actual NGS data processing
performance. If a step is highly parallelizable, and the algorithm designed for
Computing Needs for NGS Data Management and Analysis 111

it employs parallelization, then an increase in core number will most likely


lead to improved performance. On the contrary, if the step is not readily
parallelizable, or even if the task is parallelizable but the algorithm deployed
does not use parallelization, simply having more cores may not lead to
improvement in performance.

6.5 Bioinformatics Skills Required for NGS Data Analysis


For biologists and students in life sciences, acquiring basic bioinformatics
skills is greatly advantageous, as biology has become more and more data rich
and data driven. Understanding the basics of bioinformatics also facilitates
communication with bioinformaticians on the conduct of more advanced
tasks. In general, these skills include use of common computing environ-
ments, bioinformatic algorithms, and software packages. Below is a short list
of bioinformatics skills required of biologists for NGS data handling:

• Familiarity with Unix/​Linux, and the most commonly used commands


in the Unix/​Linux computing environment. This is essential to run jobs
on an HPC cluster, a local machine running a Unix/​Linux operating
system such as Ubuntu or macOS, or a Windows Subsystem for Linux
(or WSL) that provides a compatibility layer for running Linux tools
natively on Windows.
• Basic knowledge of programming languages that are commonly used
for NGS data analysis. These languages include R, Perl, and Python,
all of which are open-​source, easy to learn, and have a large user base
for help and support. While programming is not required of biologists,
understanding how an algorithm is executed step-​ by-​step can be
helpful, especially when a pre-​existing tool does not work ideally for a
special case and needs modification.
• Knowledge of key concepts in computational biology and biostat-
istics. Some computational methodologies developed in the field of
computer science, especially machine learning and data mining, have
been widely applied to high-​throughput biological data processing.
Artificial neural networks (ANNs), hidden Markov models (HMMs),
and support vector machines (SVMs) serve as good examples in this
domain. Statistical approaches such as linear and nonlinear regression
are integrated into many genomics data analysis tools and should also
be integrated into a biologist’s knowledge base.
• Basic understanding of relational database. Most of the information
currently available for the annotation and interpretation of NGS data
is captured in various databases. Knowledge of database design and
112 Next-Generation Sequencing Data Analysis

structure is the basis to extract, manipulate, and process the informa-


tion stored in these databases for generation of new biological know-
ledge. Knowing how to interact with the databases via SQL (Standard
Query Language) or APIs (Application Programmer Interfaces) is also
beneficial. The knowledge on relational databases and their operation
also helps biologists to curate, organize, and disseminate the tremen-
dous amount of information generated from NGS projects.
• Basic understanding and handling of computer hardware such as CPU,
RAM, and storage. Although strictly speaking computer hardware is
not in the realm of bioinformatics, it is nevertheless advantageous and
economical to know how to put together a workstation and put it to
use. It is also beneficial to understand how an HPC cluster, or a hetero-
geneous computing system, works through parallel processing, as NGS
tools that are designed to take advantage of these computing systems
usually function better and this knowledge can help evaluate and select
those that maximize performance built in a computing system.

For bioinformaticians who deal with NGS data, on the other hand, the
following is a list of skills that are needed:

• Proficiency with Unix-​based operating systems;


• Familiarity with a programming language such as Python, Perl, Java,
or Ruby;
• Familiarity with statistical software such as R, MATLAB, or Mathematica;
• Understanding of supercomputing, HPC (including parallel com-
puting), and enterprise data storage systems;
• Knowledge of database management languages such as MySQL or
Oracle;
• Familiarity with web authoring and web-​based user interface imple-
mentation technologies;
• Understanding of molecular biology & genetics, cell biology, and
biochemistry.

References
1. Li R, Zhu H, Ruan J, Qian W, Fang X, Shi Z, Li Y, Li S, Shan G, Kristiansen K
et al. De novo assembly of human genomes with massively parallel short read
sequencing. Genome Res 2010, 20(2):265–​272.
2. Lampa S, Dahlo M, Olason PI, Hagberg J, Spjuth O. Lessons learned from
implementing a national infrastructure in Sweden for storage and analysis of
next-​generation sequencing data. GigaScience 2013, 2(1):9.
Computing Needs for NGS Data Management and Analysis 113

3. Supernat A, Vidarsson OV, Steen VM, Stokowy T. Comparison of three variant


callers for human whole genome sequencing. Sci Rep 2018, 8(1):17851.
4. Rasche A, Lienhard M, Yaspo ML, Lehrach H, Herwig R. ARH-​seq: identi-
fication of differential splicing in RNA-​ seq data. Nucleic Acids Res 2014,
42(14):e110.
5. Farazi TA, Brown M, Morozov P, Ten Hoeve JJ, Ben-​Dov IZ, Hovestadt V,
Hafner M, Renwick N, Mihailovic A, Wessels LF et al. Bioinformatic ana-
lysis of barcoded cDNA libraries for small RNA profiling by next-​generation
sequencing. Methods 2012, 58(2):171–​187.
6. Schadt EE, Linderman MD, Sorenson J, Lee L, Nolan GP. Computational
solutions to large-​scale data management and analysis. Nat Rev Genet 2010,
11(9):647–​657.
7. Galaxy C. The Galaxy platform for accessible, reproducible and collaborative
biomedical analyses: 2022 update. Nucleic Acids Res 2022, 50(W1):W345–​W351.
8. Schatz MC, Philippakis AA, Afgan E, Banks E, Carey VJ, Carroll RJ, Culotti A,
Ellrott K, Goecks J, Grossman RL et al. Inverting the model of genomics data
sharing with the NHGRI Genomic Data Science Analysis, Visualization, and
Informatics Lab-​space. Cell Genom 2022, 2(1):100085.
115

Part III

Application-​Specific
NGS Data Analysis
7 
Transcriptomics by Bulk RNA-​Seq

7.1 Principle of RNA-​Seq
Transcriptomic analysis deals with the questions of which parts of the genome
are transcribed, and how active they are transcribed. In the past, these questions
were mostly answered with microarray, which is based on hybridization of RNA
samples to DNA probes that are specific to individual gene-​coding regions.
With this hybridization-​based approach, the repertoire of hybridization probes,
which are designed based on the current annotation of the genome, determines
what genes in the genome or which parts of the genome are analyzed, and
genomic regions that have no probe coverage are invisible. An NGS-​based
approach, on the other hand, does not depend on the current annotation of the
genome. Because it relies on sequencing of the entire RNA population, hence
the term RNA-​seq, this approach makes no assumption as to which parts of the
genome are transcribed. After sequencing, the generated reads are mapped to
the reference genome in order to search for their origin in the genome. The total
number of reads mapped to a particular genomic region represents the level of
transcriptional activity at the region. The more transcriptionally active a gen-
omic region is, the more copies of RNA transcripts it produces, and the more
reads it will generate. RNA-​seq data analysis is essentially based on counting of
reads generated from different regions of the genome.
By counting the number of reads from transcripts and therefore being digital
in nature, RNA-​seq does not suffer from the problem of signal saturation
that is observed with microarray at very high values. RNA-​seq also offers a
native capability to differentiate alternative splicing variants, which is basic-
ally achieved by detecting reads that fall on different splice junctions. While
some specially designed microarrays, like the Affymetrix Exon Arrays, can
be used to analyze alternative splicing events, standard microarrays cannot
usually make distinctions between different splicing isoforms. Also different
from microarray signals, which are continuous, raw RNA-​seq signals (i.e.,
read counts) are discrete. Because of this difference, distribution model and
methods of differential expression analysis designed for microarray data
cannot be directly applied to RNA-​seq data without modification.

DOI: 10.1201/9780429329180-10 117


118 Next-Generation Sequencing Data Analysis

7.2 Experimental Design
7.2.1 Factorial Design
Before carrying out an RNA-​seq experiment, the biological question to be
answered must be clear and well defined. This will guide experimental
design and subsequent experimental workflow from sample preparation
to data analysis. For experimental design, factorial design is usually used.
Many experiments compare transcriptomic profile between two conditions,
e.g., cancer vs. normal cells. This is a straightforward design, involving
only one biological factor (i.e., cell type). Experiments involving a single
factor may also have more than two conditions, e.g., comparison of samples
collected from multiple tissues in the body in order to detect tissue-​specific
gene expression.
If a second biological factor (e.g., treatment of a drug) is added to the
example of cancer vs. normal cell comparison, the experiment will have a
total of four (2×2) groups of samples (Table 7.1). In this two-​factor design,
besides detecting the effects of each individual factor, i.e., cell type and
drug treatment, respectively, the interacting effects between the two
factors are also detected, e.g., drug treatment may have a larger effect on
cancer cells than normal cells. If the factors contain more conditions, there
will be a total of m×n groups of samples, with m and n representing the
total number of conditions for each factor. Experiments involving more
than two factors, such as adding a time factor to the above example to
detect time-​dependent drug effects on the two cell types, are inherently
more complex and therefore more challenging to interpret, because in this
circumstance it is not easy to attribute a particular gene expression change
to a certain factor, or especially, to the interaction of these factors due to
the existence of multiple interactions (three factors involve four different
types of interactions).

7.2.2 Replication and Randomization


As with any experiment that requires proper statistical analysis, replication
and randomization is an essential component of RNA-​ seq experimental
design. Randomization refers to the random assignment of experimental

TABLE 7.1
Experimental Design Involving Two Biological Factors
Cancer Cells Normal Cells

Drug Treated Cancer +​Drug Normal +​Drug


Vehicle Treated Cancer +​Vehicle Normal +​Vehicle
Transcriptomics by Bulk RNA-Seq 119

subjects or targets into each group. This is to avoid introducing unwanted


biases to the sample collection process. To generalize the gene expression
differences observed from groups of samples to the respective populations,
within-​group variability in the expression of each gene has to be estimated,
which requires replication. To meet this requirement at least three replicates
need to be included within each group. The more replicates each group has,
the more accuracy there is in within-​group biological variability estimation,
and therefore the more confidence to call a gene differentially expressed.
While differential gene expression can be detected from unreplicated data, the
results are limited to the tested samples and not easily generalizable. Due to
the lack of knowledge on biological variation within each group, it is unreal-
istic to draw conclusions on the population from an unreplicated experiment.

7.2.3 Sample Preparation and Sequencing Library Preparation


Since gene expression is highly plastic and varies greatly with internal
(such as tissue and cell type, developmental stage, circadian rhythm, etc.)
and external (such as environmental stress) conditions, samples should
be collected in a way that minimizes the effects of irrelevant factors. If the
influence of such factors cannot be totally avoided, they should be balanced
across groups. As many biological samples contain different cell types, this
heterogeneity in cell composition is another factor that may confound data
interpretation (potential heterogeneity in cell composition can be revealed by
single-cell RNA-​seq, to be covered in the next chapter). Use of homogeneous
target cells is preferred whenever possible as this will greatly improve data
quality and experimental reproducibility.
To prepare samples for RNA-​seq, total RNA is first extracted from samples
of contrasting conditions. High-​quality RNA extraction is essential to obtain
quality RNA-​seq data. The leading cause of low-​quality RNA is degrad-
ation. To detect the intactness of RNA molecules in samples, quality metrics
such as RNA Integrity Number (or RIN) are often used. RIN is generated
using a neural network-​based algorithm from Bioanalyzer fragment size pro-
file of an RNA sample, having values from 1 to 10 with 10 being the best
possible sample quality. One prerequisite to extracting high-​quality RNA
is to snap-​freeze tissue samples whenever possible to avoid potential RNA
degradation. Under circumstances where this is not possible (e.g., sample
collection in the field), RNA stabilizing reagent (such as RNAlater) can be
used. For RNA samples prepared under certain circumstances such as those
from historical samples or formalin-​fixed paraffin-​embedded (FFPE) clinical
tissues, however, RNA degradation can be unavoidable. Even from highly
degraded RNA samples such as these, useful data may still be generated
with the use of RNA-​seq library construction strategies that are more tol-
erant of RNA degradation [1]. Among other issues that might affect RNA-​seq
data quality is genomic DNA contamination. To remove DNA contaminants,
a DNase treatment step during RNA extraction is recommended. It should
120 Next-Generation Sequencing Data Analysis

also be noted that many RNA extraction protocols do not retain small RNA
species including miRNAs. If these species are also of interest (more on small
RNA sequencing in the Chapter 9), alternative protocols (such as the TRIzol
method) need to be used.
Besides quality, the quantity of RNA sample available also determines
RNA-​seq library prep strategy. With enzyme engineering and as a result
improvement in library construction chemistry, preparing sequencing
libraries from increasingly small quantities of RNA is no longer a barrier.
This usually involves signal amplification in order to produce enough library
molecules for sequencing. While the needed amplification can introduce bias
to the process, its impact on the detection of differential gene expression has
been found to be limited [2]. The greatly increased sensitivity in RNA-​seq
library making has also made sequencing of transcripts from a single cell a
reality (see next chapter for single-cell RNA-​seq).
There are two general approaches to constructing RNA-​seq sequencing
libraries. One approach is based on direct enrichment of mRNA molecules,
the major detection targets for the majority of RNA-​ seq work. Because
most eukaryotic mRNAs have a poly-​A tail (Chapter 3, Section 3.3.4), this
approach is carried out through the use of poly-​T capture probes to enrich for
mRNA molecules carrying such a tail. The other approach is based on deple-
tion of ribosomal RNAs (rRNAs), since rRNAs are usually the predominant
but uninformative component in total RNA extractions. Depletion of rRNAs
is typically based on hybridization using rRNA-​specific probes, followed
by their capture and subsequent removal. Other rRNA depletion strat-
egies include degradation by duplex-​specific nuclease (DSN), which relies
on denaturation-​reassociation kinetics to remove extremely abundant RNA
species including rRNAs [3], and RNase H selective depletion, on the basis of
binding rRNAs with rRNA-​specific DNA probes and then using RNase H to
digest bound rRNAs. Library prep based on the rRNA depletion approach is
more tolerant of RNA degradation issues.
After mRNA enrichment or rRNA depletion, subsequent RNA sequencing
library preparatory process typically involves reverse transcription to cDNA
using random primers, followed by fragmentation (for short-read platforms)
and attachment of sequencing adapters. This sequencing library construction
process may also introduce bias to the subsequent sequencing and data gen-
eration. For example, the use of poly-​T based mRNA enrichment introduces
3’ end bias, as this procedure precludes analysis of those mRNAs and other
non-​coding RNAs that do not have the poly-​A tail structure [4]. If these RNA
species are of interest, a library prep process based on the use of rRNA deple-
tion can be employed.
Compared to short-read sequencing, long-read sequencing offers new
RNA-​seq capabilities and options that are impossible or difficult to per-
form using short-read sequencing. For example, both ONT and PacBio
platforms provide full-​ length transcript sequencing, and thereby enable
Transcriptomics by Bulk RNA-Seq 121

characterization and quantification of gene expression at the splicing isoform


level. Besides sequencing cDNA, ONT sequencing can also sequence RNA
molecules directly that removes some of the biases introduced during cDNA-​
based library construction.

7.2.4 Sequencing Strategy
Sequencing depth and read length are two major factors to consider when
sequencing bulk RNA-​seq libraries especially with short-read sequencing.
The factor of sequencing depth, that is, how many reads to obtain, is based
on a number of factors, mainly the size of the organism’s genome, the pur-
pose of the study, and ultimately statistical rigor (effect size and statistical
power). Small genomes, such as those of bacteria, require less reads to ana-
lyze than large genomes, such as those of mammalian species. If the pur-
pose of a study is to identify differentially expressed genes among those
expressed at intermediate to high abundance levels, it requires fewer reads
than studies that aim to encompass low-​abundance genes, study alterna-
tive splicing events, or discover new transcripts. As a general guideline,
for a gene expression profiling experiment targeting intermediate-​to high-​
abundance transcripts, a sequencing depth of 5–​25 million reads, depending
on the size the genome, is suggested. To cover transcripts of lower abundance
or common alternative splicing variants, 20–​50 million reads are suggested.
For more thorough coverage of the transcriptome and/​or discovery of new
transcripts, 100–​300 million reads are often needed. As for sequencing read
length, for gene expression profiling, single end 50–​75 bp reads are typically
long enough to map to their originating genes in the genome. For assembly of
new transcripts and/​or identification of alternative splicing isoforms, longer
and often paired-​end sequencing reads, such as paired-​end 150 bp reads, are
often acquired.
The number of sample replicates also largely affects the detection power of
an RNA-​seq study, as sample replication provides estimation on gene expres-
sion dispersion across biological subjects within a group. While a minimum
of three biological replicates is commonly used, specially designed RNA-​seq
power analysis tools can be used to calculate sample size to achieve a detec-
tion power. These tools, including Scotty [5], ssizeRNA [6], PROPER [7], and
RnaSeqSampleSize [8], are designed based on different statistical models.
For example, while PROPER and RnaSeqSampleSize are based on negative
binomial model, Scotty and ssizeRNA use Poisson-​lognormal and linear
models, respectively. As to be covered more later in this chapter (Section
7.3.3 “Identification of Differentially Expressed Genes”), these models pro-
vide different approximations to RNA-​seq data distribution. Sample size
calculations using these tools require as input a number of parameters,
including the total number of genes expressed, the percentage of genes
expected to be differentially expressed, the minimal fold change needed to
122 Next-Generation Sequencing Data Analysis

call differential expression, false discovery rate, average read count (related
to sequencing depth), and the desired statistical power. Because most of the
parameters are not known a priori, recently published data collected from
similar conditions may be used to provide some guidance. To start on a
species or cell type that has not yet been studied, it might be useful to try out
a small number of samples first to get a general idea on the composition of the
target transcriptome and the variability between biological replicates. Besides
detection power, experimental and sequencing costs are also key factors in
deciding sample size and sequencing depth. For projects on a budget, it has
been reported that increasing the number of biological replicates is more
effective in boosting detection power than increasing sequencing depth [9].
Besides sequencing depth and read length, other considerations when
planning for sequencing samples include how to arrange samples on a
sequencer in terms of flow cell or lane assignment. Here a balanced block
design [10] should be used to minimize technical variation due to flow
cell-​to-​flow cell or lane-​to-​lane difference. In such a design, samples from
different conditions are multiplexed on the same flow cell(s) or lanes,
instead of running different samples or conditions on separate flow cell(s)
or lanes.

7.3 RNA-​Seq Data Analysis


7.3.1 Read Mapping
The first step after an RNA sequencing run is to examine the run summary
with regard to the total number of reads generated, quality score distribution,
GC content, and other indices of the sequencing run as detailed in Chapter 5.
Based on such QC results, reads filtering and base trimming can be conducted
to remove low-​quality reads or basecalls. The distribution of sequencing
depth across samples should also be checked to ensure all samples receiving
expected numbers of reads. In addition, there are tools such as subSeq [11]
that can be used to determine whether the desired detection power is reached
and whether additional sequencing would lead to significant increase in
detection sensitivity.
The subsequent mapping of RNA-​seq reads to a reference genome is in
general more complex than the general read mapping procedure described
in Chapter 5. Because eukaryotic mRNAs are generated from splicing out of
introns and joining of exons, many RNA-​seq reads may not map contigu-
ously to the reference genomic sequence. As prokaryotic mRNA generation
does not involve splicing, the aligners introduced in Chapter 5 for contiguous
(ungapped) mapping, such as Bowtie and BWA, can still be used. Mapping of
eukaryotic RNA-​seq reads, however, creates a challenge to these aligners. To
Transcriptomics by Bulk RNA-Seq 123

address this challenge, two approaches have been developed. One is to use the
current gene exonic annotation in the reference genome to build a database of
reference transcript sequences that join currently annotated exons. RNA-​seq
reads are then searched against this reference transcripts database using the
ungapped read aligners. Examples of annotation-​guided mappers are RUM
[12] and SpliceSeq [13]. These mappers may produce better outcomes when
high accuracy and reliability are emphasized. This approach, however, does
not provide the capability to discover novel transcripts. In addition, it leads
to high rate of multi-​mapping, as a read that maps to common exon(s) shared
by multiple splicing isoforms of a gene is counted multiple times.
The other approach conducts ab initio splice junction detection, and there-
fore does not depend on genome annotation. Depending on their method-
ology, ab initio spliced mappers can be classified into two categories: methods
using “exon-​first” and those using “seed-​and-​extend.” The exon-​first methods
include TopHat/​TopHat2 [14, 15], MapSplice [16], SpliceMap [17], and GEM
[18]. They first align reads to a reference genome to identify unspliced con-
tinuous reads (i.e., exonic reads first), and then predict splice junctions
out of the initially unmapped reads based on the initial mapping results.
Taking TopHat/​TopHat2 as an example, they first use Bowtie/​Bowtie2 to
align reads to the reference genome. Reads that map to the reference con-
tinuously without interruption are then clustered based on their mapping
position. The clusters, supposedly representing exonic regions, are used to
search for splicing junctions from the remaining reads. The seed-​and-​extend
methods, on the other hand, use part of reads as substrings (or k-​mers) to
initiate the mapping process, followed by extension of candidate hits to
locate splicing sites. Examples of methods in this category include STAR [19],
HISAT/​HISAT2 [20, 21], and GMAP/​GSNAP [22]. Among these methods,
STAR employs a two-​step process for gapped alignment. The first is a seed
searching step, aiming to sequentially locate substrings of maximum length
from a read that each matches exactly to one or more substrings in the refer-
ence genome. If this step does not reach the end of the read due to the presence
of mismatches, it will use the located seed region(s) as anchors to extend
the alignment. In the second step, alignment of the entire read sequence is
built by joining all the seed regions located in the first step. HISAT applies an
algorithm called hierarchical indexing to achieve splice-​aware alignment. It
starts with a global search using FM indexing of the whole reference genome
to identify the genomic location(s) of a read using part of its sequence as
seed. Such a location is then used as an anchor to extend the alignment. Once
the alignment cannot be extended further, e.g., reaching a splicing junction,
a local search is then performed using FM indexing of the local region to
map the rest of the read (Figure 7.1). A hybrid strategy combining the two
is also used sometimes, with the exon-​first approach employed for mapping
unspliced reads and the seed-​and-​extend approach for spliced reads. As they
do not rely on current genomic annotations, these ab initio methods are suit-
able to identify new splicing events and variants.
124 Next-Generation Sequencing Data Analysis

FIGURE 7.1
Mapping of RNA-​seq reads with HISAT. Three representative reads are shown on the top, i.e.,
one exonic read (1), one read spanning a splice junction with a short anchor in one exon (2), and
another junction-​spanning read with long anchor in each exon (3). Panel (a) shows alignment
of Read1 with a global FM genome index search using partial read sequence, followed by an
extension step to align the rest of the read. Panel (b) show that when the global search and
extension are halted at the junction, a local search using the region’s FM index is performed to
align the remaining short sequence. Panel (c) shows that to align Read 3, a second extension
step is conducted after the local FM index search. The shown exemplary reads are error-​free
and 100 bases in length. (Adapted by permission from Springer Nature Customer Service
Centre GmbH: Springer Nature, Nature Methods, HISAT: a fast spliced aligner with low memory
requirements, Kim, D., Langmead, B. & Salzberg, S., Copyright 2015.)
Transcriptomics by Bulk RNA-Seq 125

To map long RNA-​seq reads generated on the PacBio and ONT platforms,
some of the short-​read mappers introduced above can still be used, such
as GMAP [23]. More commonly, however, this task is performed using tools
specially designed for long reads. For example, minimap2 (introduced in
Chapter 5) has a splice-​aware option for mapping long RNA sequencing
reads. Other currently available tools include deSALT [24], GraphMap2 [25],
and uLTRA [26].
The percentage of reads that are mapped to the genome is an important
QC parameter. While it is variable depending on a number of factors such
as aligning method and species, this number usually falls within the range
of 70–​90%. The percentage of reads that map to rRNA regions is dependent
on and a measure of the efficiency of the rRNA depletion step. Due to tech-
nical and biological reasons, it is usually impossible to remove all rRNA
molecules. The percentage of rRNA reads can vary greatly, from 1–​2% to
35% or more. For downstream analysis rRNA reads are filtered out so they
do not usually affect subsequent normalization. Duplicate reads, a common
occurrence in an RNA-​seq experiment, can be caused by biological factors,
such as over-​presentation of a small number of highly expressed genes, and/​
or technical reasons, such as PCR over-​amplification. It is possible to have
a high percentage of duplicate reads, e.g., 40–​60%, in a run. While it is still
debatable as to how to treat duplicate reads, because of the biological factors
involved in their formation they should not be simply removed. Some experi-
mental approaches, such as removing some of the highly expressed genes
prior to library construction, or using paired-​end reads, can help reduce the
number of duplicate reads. With regard to genomic coverage, RNA-​seq QC
tools, including RNA-​SeQC 2 [27], RSeQC [28], and QoRTs [29], report on
the percentage of reads that are intragenic, that is, those that map within
genes (including exons or introns), or intergenic, for those that map to gen-
omic space between genes. These tools also report other data quality metrics,
including percentage of total aligned reads, percentage of rRNA reads, as
well as rate of duplicate reads.
If the species under study does not have a sequenced reference genome
against which to map RNA-​seq reads, two approaches exist. One is to map the
reads to a related species that has a reference genome, while the alternative is
to assemble the target transcriptome de novo. The de novo assembly approach
is more computationally intensive, but it does not rely on reference genomic
sequence. Currently available de novo transcriptome assemblers include
rnaSPAdes [30], Trinity [31], Bridger [32], Trans-​ABySS [33], SOAPdenovo-​
Trans [34], Oases [35], and StringTie/​ StringTie2 [36, 37]. Among these
assemblers rnaSPAdes and StringTie2 can be used with long RNA reads,
or a hybrid of long and short reads [38, 39]. These de novo assemblers are
suited when no related species or only very distantly related species with
a reference genome exists, or the target genome, despite with available ref-
erence sequence, is heavily fragmented or altered (such as in tumor cells).
It should also be noted that if a related reference genome exists with 85%
126 Next-Generation Sequencing Data Analysis

or higher sequence similarity with the species under study, mapping to the
related genome may work equally well, or even better, compared to the
de novo assembly approach. This is especially true when studying alterna-
tive splicing variants. These de novo approaches are also applicable to cases
where aberrant transcripts are expected, or novel transcripts are the detec-
tion targets. For these de novo assembly approaches, paired-​end sequencing
or long-read sequencing are more advantageous compared to single-​end
short reads.

7.3.2 Quantification of Reads
After mapping of reads, the number of reads mapped to each gene/​tran-
script needs to be counted to generate a table with rows representing genes/​
transcripts and columns different samples. Such an expression matrix is the
basis for subsequent differential expression determination. This read quan-
tification process can be performed using tools such as featureCounts as part
of Subread [40], htseq-​count as distributed with the HT-​Seq Python frame-
work [41], RSEM [42], eXpress [43], or Cufflinks [44]. Among these tools,
featureCounts and htseq-​count are read count-​based, requiring as input
SAM/​BAM alignment files and a genomic feature annotation GFF/​GTF file.
They generally discard reads that map to multiple regions in the genome
or overlap multiple genomic features. RSEM, eXpress, and Cufflinks, on the
other hand, are model-​based, requiring as input SAM/​BAM alignments as
well as a transcriptome reference file containing transcript sequences. They
assign multi-​mapped or ambiguous reads to transcripts based on prob-
ability from the use of the expectation-​maximization algorithm. Because
of the differences in how they quantify genes/​transcripts, the selection
of counting methods has been shown to have an effect on quantification
results [45].
Gene/​ transcript expression can also be quantified without mapping
reads to a reference genome or transcriptome. Examples of such mapping-​
independent algorithms include kallisto [46], Salmon [47], and Sailfish [48].
These methods rely on pseudo-​or quasi-​alignment of k-​mers extracted
from reads, instead of the entire reads, to achieve transcript quantifica-
tion at vastly faster speed. For example, kallisto is a pseudoaligner that
aligns k-​mers in reads to a hash table built from k-​mers that represent
different transcripts in the reference transcriptome. Although not relying
on mapping of entire reads, this method enables rapid determination of the
compatibility of reads with target transcripts, as it preserves the key infor-
mation needed for transcript quantification. Since only k-​mers need to be
aligned to the hash table, the speed is greatly increased with similar quan-
tification performance to the mapping-​based methods above. They perform
well on highly expressed protein-​coding genes but less so on rare or short
transcripts [49].
Transcriptomics by Bulk RNA-Seq 127

7.3.3 Normalization
As mentioned previously, the basic principle of determining gene expres-
sion levels through RNA-​seq is that the more active a gene is transcribed,
the more reads we should be able to observe from it. To apply this basic prin-
ciple to gene expression quantification and cross-​condition comparison, at
least two factors must be taken into consideration. The first is sequencing
depth. If a sample is split into two halves, and one half is sequenced to a
depth that is twice of that of the other, for the same gene the former will
generate twice as many reads as the latter although both are from the same
sample. The other factor is the length of gene transcript. If one gene tran-
script is twice the length of another gene transcript, the longer transcript
will also produce twice as many reads as the shorter one. Because of these
confounding factors, prior to comparing abundance of reads from different
genes across samples in different conditions, the number of reads for each
gene needs to be normalized against both factors using the following formula
to ensure different samples and genes can be directly compared,

gi , j × SF
ei , j =
ai × l j

where ei,j is the normalized expression level of gene j in sample i, gi,j is the
number of reads mapped to the gene in the same sample, ai is the total
number of mapped reads (depth) in sample i, and lj is the length of gene j. SF
is a scaling factor and equals to 109 when ei,j is presented as RPKM or FPKM
(Reads, or Fragments [for paired-​end reads], per Kilobase of transcript per
Million mapped reads).
The calculation of RPKM or FPKM is the simplest form of RNA-​seq data
normalization. In a nutshell, normalization deals with non-​intended factors
and/​or technical bias, such as those that lead to unwanted variation in total
read counts in different samples. By correcting for the unwanted effects of
these factors or bias, the normalization process puts the focus on the biological
difference of interest, and makes samples comparable. Since the introduction
of RKPM or FKPM as an early normalization approach for RNA-​seq data,
other methods of normalization have also been developed. Some of these
methods employ a similar strategy to adjust for sequencing depth. This group
of methods normalize RNA-​seq data through dividing gene read counts by
either (1) the total number of mapped reads (i.e., the total count approach);
(2) the total read count in the upper quartile (the upper quartile approach)
[50]; and (3) the median read count (the median approach). Another method
called quantile normalization sorts gene read count levels and adjusts quan-
tile means to be the equal across all samples, so that all samples have the
same empirical distribution [51]. These sequencing depth-​based methods do
not normalize against gene length, as it is not needed if the goal is to detect
128 Next-Generation Sequencing Data Analysis

relative expression changes of same genes between groups, rather than com-
pare relative abundance levels of different genes in the same samples.
Subsequently, more sophisticated normalization approaches are developed
based on the assumption that the majority of genes are not differentially
expressed, and for those that show differential expression, the proportion of
up-​and down-​regulation is about equal. These approaches include those that
are employed by two commonly used RNA-​seq analysis tools, DESeq2 [52]
and edgeR [53]. DESeq2 employs a method called relative log expression, or
RLE, which is essentially carried out through dividing the read count of each
gene in each sample by a scaling factor. To compute the scaling factor for each
sample, the ratio of each gene’s read count in a sample over its geometric
mean across all samples is first calculated. After calculating this ratio for all
genes in the sample, the median of this ratio is used as the scaling factor. The
edgeR package employs a approach called Trimmed Means of M-​values, or
TMM [54]. In this approach, one sample is used as the reference and others
as test samples. TMM is computed as the weighted mean of gene count log
ratios between a test sample and the reference, excluding genes of highest
expression and those with the highest expression log ratios. Based on the
assumption of no differential expression in the majority of genes, the TMMs
should be 1 (or very close to 1). If not, a scaling factor should be applied
to each sample to adjust their TMMs to the target value of 1. Multiplying
the scaling factor with the total number of mapped reads generates effective
library size. The normalization is then carried out through dividing raw
reads count by the effective library size, i.e., normalized read count =​raw
read count ⁄ (scaling factor × total number of mapped reads).
Among other approaches are those that use iterative processes to achieve
normalization, as exemplified by TbT [55], DEGES [56], and PoissonSeq [57].
Based on the same assumption that there is no overall differential expres-
sion, these methods use a multi-​iteration process. For example, DEGES,
or Differentially Expression Genes Elimination Strategy, uses a process
to repeatedly remove potential differential genes until their elimination,
prior to calculating the final normalization factor. It starts with using any
of the normalization methods introduced above, e.g., TMM, followed by a
test for differential expression using a differential detection method (to be
introduced next). After removal of the DE genes, the same process is repeated
until convergence.
There are also normalization methods that use a list of housekeeping genes
or spike-​in controls as normalization standard. The use of housekeeping
genes or spike-​in controls is for conditions in which the assumption that
the majority of genes are not differentially expressed might be violated. In
this approach, a set of constitutively expressed housekeeping genes that are
known to stay unchanged in expression under the study conditions, or a
panel of artificial spike-​in controls that mimic natural mRNA and are added
to biological samples at known concentrations, is used as the basis against
which other genes are normalized.
Transcriptomics by Bulk RNA-Seq 129

7.3.4 Batch Effect Removal


Normalization cannot fully address signal variation caused by collection of
samples in batches. While it is ideal to collect all samples at once using a
balanced experimental design, due to logistical or realistic considerations
samples are sometimes collected in different batches that may involve
different personnel, instrument, or protocol. Data collected in batches may
introduce unwanted, non-​biological variation in the RNA-​seq signal, which
will confound downstream analysis if not removed. Principal components
analysis (PCA) can be used as an intuitive tool to detect batch effects, as it
provides a convenient way to visualize relative relationships among samples,
and samples are expected to cluster together based on experimental design
(Figure 7.2). Tools such as BatchQC [58] can also be used to detect potential
batch effects. Once batch effects are detected, specially designed tools, such
as ComBat [59], RUVSeq [60], and svaseq [61], can be used to remove them.
Among these tools, ComBat uses an empirical Bayes approach to adjust for
batch effects, making it robust for data collected from batches with small
sample sizes. ComBat-​seq is an adaptation of the original ComBat for RNA-​
seq count data, including the use of a negative binomial regression model
[62]. Besides removing batch effects from a single experiment, these tools can
also be employed for integrated analysis of large datasets collected from mul-
tiple experiments or groups to boost detection power.

7.3.5 Identification of Differentially Expressed Genes


To compare gene expression in different groups and identify DE genes, the
distribution model of the data needs to be established first in order to decide
on the appropriate statistic tests to be used. While microarray data can be
treated as normally distributed variables after log transformation, the RNA-​
seq read count values, being discrete in nature, cannot be approximated by
continuous distributions even after transformation. In general, count data,
including the RNA-​ seq data, follows the Poisson distribution, which is
characterized by the mean of the distribution being equal to the variance.
While this distribution can be and has been used to model RNA-​seq data [50,
63], it has also been observed that in RNA-​seq data genes with larger mean
counts tend to have greater variance, causing the over-​dispersion problem
[64] (see Figure 7.3). To deal with this problem, an over-​dispersed Poisson
process, or as an approximation the negative binomial distribution, is often
applied. Other distribution models that have been used in RNA-​seq data
analysis tools, including the Poisson log-​linear model used by PoissonSeq
[57] and the log-​normal model used by limma [65] and Ballgown [66], have
also been found to perform well under many circumstances.
On the identification of DE genes based on these models, there is a list
of methods (Table 7.2) to choose from, among which the commonly used
ones are baySeq [67], Cuffdiff 2 [68], DEGseq [69], DESeq2, EBSeq [70], and
130 Next-Generation Sequencing Data Analysis

FIGURE 7.2
Removal of batch effects using ComBat-​seq. PCA plots are shown before (A) and after (B) batch
effects correction. In this example, three batches of breast cancer tissue samples that overexpress
three genes (HER2, EGFR, and KRAS) separately with their controls expressing GFP are shown.
The correction effectively removes batch effects on the control samples from the three batches,
while maintains the effects of the transgenes. (Adapted from Y Zhang, G Parmigiani, ComBat-​
seq: batch effect adjustment for RNA-​seq count data, NAR Genomics and Bioinformatics 2020,
2(3):lqaa078.)

edgeR [53]. While DEGseq has been developed based on the Poisson distri-
bution, baySeq, Cuffdiff 2, DESeq2, EBSeq, and edgeR have been designed
on the negative binomial distribution. To detect DE genes, these packages
use different approaches. For example, baySeq and EBSeq employ empirical
Transcriptomics by Bulk RNA-Seq 131

FIGURE 7.3
The overdispersion problem in RNA-​seq data. Poisson distribution is often used to model
RNA-​seq data, but instead of the variance/​dispersion being approximately equal to the mean
as assumed by the distribution, the variance in RNA-​seq data is often dependent on the mean.
The purple line represents the relationship between variance and mean based on the Poisson
distribution, while the solid and dashed orange lines represent local regressions used by DESeq
and edgeR, respectively, based on negative binomial distribution. (Modified from Anders
S. and Huber W. (2010) Differential expression analysis for sequence count data. Genome
Biology, 11, R106. Used under the terms of the Creative Commons Attribution License [http://​
creativecommons.org/​licenses/​by/​2.0] © 2010 Anders et al.)

TABLE 7.2
Tools for Detection of DE Genes
Name Description Reference

DESeq2 Employs negative binomial generalized linear modeling and Wald [52]
test to detect DE genes. Uses empirical Bayes estimation to shrink
dispersions and fold changes to increase detection stability
edgeR Detects DE genes based on negative binomial distribution, using [53]
techniques including exact test, generalized linear modeling,
quasi-​likelihood F-​test, and empirical Bayes
limma Fits a gene-​based linear model for DE analysis. Originally [65]
developed for microarray gene expression data, it uses the voom
function for RNA-​seq data to apply empirical Bayes to estimate
gene-​wise variability
NOISeq A nonparametric and data-​adaptive method that detects DE genes [72]
based on simultaneous comparison of fold change and absolute
expression difference
(continued)
132 Next-Generation Sequencing Data Analysis

TABLE 7.2 (Continued)


Tools for Detection of DE Genes
Name Description Reference

Cuffdiff 2 Combines estimates of count uncertainty and overdispersion to [68]


model fragment count variability, which is then used to test for
significant DE changes
EBSeq Applies empirical Bayesian methods to identify DE genes and [70]
splicing isoforms
baySeq Builds on negative binomial distribution, and uses an empirical [67]
Bayesian approach to estimate posterior probabilities for each of a
set of models to determine DE
SAMseq Another nonparametric method originally designed for microarray [71]
data adapted for RNA-​seq data. Uses resampling to account for
differences in sequencing depth when performing DE analysis

Bayesian based methods, in which two alternative models are proposed


for each gene, with one assuming differential expression and another
assuming null. Given the observed counts, the posterior likelihood for the
differential expression model is used to identify DE genes. Cuffdiff 2 uses
the T statistic, which equals the ratio of mean(log[y]‌) over variance(log[y]),
with y representing the expression ratio of a gene between two groups.
Since this statistic approximately follows a normal distribution, t-​test is
used for DE analysis. DEGseq employs several methods to identify DE
genes, including methods based on the MA-​plot, Fisher’s exact test, likeli-
hood ratio test, and samrWrapper (a wrapper of functions in SAM,
which is originally designed for identifying differential gene expression
from microarray data). DESeq2 applies a generalized linear model (GLM)
approach to identify DE genes with a Wald test. Another commonly used
method, edgeR, tests for differential gene expression using an exact test
that is highly parallel to Fisher’s exact test for experiments with one factor,
and GLM likelihood ratio test or quasi-​likelihood F-​test for general multifac-
torial experiments.
Packages that are not based on the Poisson distribution or negative bino-
mial distribution are also used for differential expression analysis. For
example, limma uses linear modeling after logarithmic transformation
of read counts, and a moderated t-​statistic to find DE genes. PoissonSeq
conducts DE analysis based on a test of a correlation term between gene and
experimental conditions, which follows a chi-​squared distribution model.
The adaptation of SAM for RNA-​seq data analysis has led to the develop-
ment of SAMseq, which, different from the original SAM, is based on a non-​
parametric approach [71]. NOISeq provides another example of using the
non-​parametric approach for cases where a probability distribution model of
the data may not hold [72]. Despite the differences in the statistical models
used, benchmarking studies have shown that many of the tools introduced
Transcriptomics by Bulk RNA-Seq 133

above generate similar results for well-​powered studies, and those based on
the non-​parametric approach are often equally effective [73, 74].
Most of the currently available methods are designed to handle samples
with biological replicates. Under non-​ideal circumstances when RNA-​seq is
performed without replication, it becomes impossible to estimate biological
variability for a satisfactory statistical analysis. For such cases, the only indi-
cator of differential expression is fold change. Some of the above tools can still
take such data, such as edgeR, which offers an option to manually input a dis-
persion value estimated from similar studies, and NOISeq, which provides
technical replicates through simulation of the data assuming a multinomial
distribution, acting as alternative means to estimate biological variability for
a significance analysis. There are also tools especially designed for RNA-​seq
experiments without replication, including GFOLD [75] and ACDtool [76].
ACDtool is an implementation of the method originally proposed by Audic
and Claverie developed on the basis of Poisson distribution [77]. Although
originally designed for analyzing relatively small data sets (<10 K reads),
the A-​C statistic and its implementation through ACDtool is equally sensi-
tive and applicable to the much larger NGS data sets that contain millions of
reads without replicates.

7.3.6 Multiple Testing Correction


A typical RNA-​seq experiment involves a small number of samples, but a
large number of genes, to test for differential expression between conditions.
If only one gene is compared, a p of 0.05 is usually used as a threshold for
type I error (false positive), i.e., accepting that there is a 5% chance of calling
a gene differentially expressed while it is not. When a large number of genes
are compared simultaneously, however, using the same p of 0.05 for each
gene will lead to a significant number of false discoveries. For example, if
10,000 genes are compared, at p =​0.05 there will be 500 genes to be called
differentially expressed just by random chance. Therefore, an adjustment is
needed to correct for the high rate of false discoveries caused by multiple
testing. One adjustment approach is to divide the p by the total number
of genes/​comparisons being conducted, e.g., using an adjusted p of 5x10-​6
when comparing 10,000 genes. This approach, called Bonferroni correction,
controls the family wise error rate (FWER), i.e., the probability of detecting
one or more false-​positive genes. While it is straightforward to implement, it
is very conservative and often too stringent, leading to false negatives (also
called type II errors).
A more practical and powerful approach was proposed by Benjamini and
Hochberg (1995) [78]. Instead of controlling FWER, this approach controls
the proportion of false positives among identified genes, i.e., the false dis-
covery rate (FDR). To illustrate, when using an FDR of 0.05, we accept the
probability that 5% of the genes detected to be differentially expressed are
134 Next-Generation Sequencing Data Analysis

false positives. As an example, if 1,000 genes are identified to be differen-


tially expressed, 50 genes are expected to be false positives by chance. FDR-​
adjusted p values are often called q values. If a gene has a q value of 0.05, it
means that 5% of the genes with q values less than 0.05 are expected to be
false positives. This approach, through relaxing the expectation and allowing
a small proportion of false positives among all discoveries, leads to a gain in
detection power. All DE detection tools introduced above offer options for
multiple testing correction.

7.3.7 Gene Clustering
Genes showing differential expression pattern between conditions can be
grouped into different clusters based on their overall expression pattern.
This unsupervised process helps uncover different patterns of overall gene
expression changes, and thereby serves as an important exploratory step to
find key target genes for further investigation and hypothesis generation.
Among the most widely used clustering algorithms are hierarchical and k-​
means clustering. Hierarchical clustering aims to build a dendrogram based
on similarity of expression between genes. This clustering method can take
either a “bottom-​up,” also called agglomerative, approach in which each gene
is in their own cluster initially and then recursively merged until only one
cluster remains, or a “top-​down” (divisive) process that employs a reverse
process. For RNA-​seq data, the agglomerative approach is used more com-
monly. Besides clustering genes, samples are often clustered at the same time
to uncover relationships among them (Figure 7.4). With k-​means clustering,
the number of clusters, i.e., the k value, needs to be defined a priori. Performing
hierarchical clustering first can help assess what k value to use. The objective
of k-​means clustering is to assign each gene to the nearest cluster mean. For
both hierarchical and k-​means clustering, an often-​used similarity measure is
Pearson correlation coefficient. Besides these two commonly used clustering
algorithms, other clustering methods include Self-​Organizing Map (SOM)
and Partitioning Around Medoids (PAM). These clustering processes can be
performed using functions in R such as “hclust” for hierarchical clustering
and “kmeans” for k-​means clustering.

7.3.8 Functional Analysis of Identified Genes


Besides clustering, functional analysis of identified DE genes is also necessary
to connect the genes, usually in large numbers, to the biological question
under study. This analysis can be conducted at multiple levels, including
Gene Ontology (GO), biological pathway, and gene network. GO provides
hierarchically structured, standardized terms to describe biological processes,
molecular functions, and cellular components associated with a gene [79].
A biological pathway, on the other hand, refers to a series of molecular
reactions that leads to a cellular event or product. Commonly used pathway
Transcriptomics by Bulk RNA-Seq 135

FIGURE 7.4
Hierarchical clustering of DE genes as well as experimental samples. RNA-​seq data shown here
is collected from cultured fibroblasts that were subjected to two treatment conditions (irradiation
[IR] and TGF-​β1 [TGF-​β], in comparison with control [CTR]). (From Mellone M, Hanley CJ,
Thirdborough S, Mellows T, Garcia E, Woo J, Tod J, Frampton S, Jenei V, Moutasim KA, Kabir TD,
Brennan PA, Venturi G, et al. (2016) Induction of fibroblast senescence generates a non-​fibrogenic
myofibroblast phenotype that differentially impacts on cancer prognosis. Aging (Albany NY).
159:114–​132. Used under the terms of the Creative Commons Attribution License (CC BY 3.0)
[https://​crea​tive​comm​ons.org/​licen​ses/​by/​3.0/​] © 2016 Mellone et al.)

databases include KEGG [80], Pathway Commons [81], WikiPathways


[82], and Reactome [83]. Tools such as Enrichr [84], GOseq [85], GOrilla
[86], g:Profiler [87], DAVID (or Database for Annotation, Visualization and
Integrated Discovery) [88], and ToppGene [89] detect enrichment of GO terms
and biological pathways in identified genes. The statistical significance of
136 Next-Generation Sequencing Data Analysis

this enrichment is usually calculated using the hypergeometric distribution,


or Fisher’s exact test. An alternative approach is the Gene Set Enrichment
Analysis (GSEA), which instead of using a filtered list of genes, uses the
entire gene set for functional analysis [90]. Not relying on a somewhat arbi-
trary cutoff for gene selection, the GSEA approach increases sensitivity of the
analysis and can pick up weaker signals that might be otherwise missed.
For gene regulatory network analysis, tools like Cytoscape or Ingenuity
Pathway Analysis (IPA, commercial) are often used. Gene network can be
reconstructed on the basis of currently available experimental evidence, or
co-​expression patterns. Cytoscape, for example, provides a range of apps
for gene network analysis and visualization, such as stringApp [91] and
GeneMANIA [92], which provide gene-​gene (or protein-​protein) interaction
information based on experimental evidence catalogued in databases such as
BioGrid, Integrated Interactions Database, and STRING. In addition, some
Cytoscape Apps, such as ClueGO [93] and EnrichmentMap [94], offer visual-
ization of gene set enrichment results (e.g., those from GSEA) as a network.

7.3.9 Differential Splicing Analysis


Besides overall expression level changes, eukaryotic genes also undergo alter-
native splicing to produce different isoforms of transcripts (see Chapter 3). As
differential splicing may exist even in the absence of overall gene expression
level changes, analysis of differential splicing (DS) adds another dimension
to transcriptomic profiling. DS analysis methods can be generally classified
into different categories based on detection target (exon, splicing event, or
isoform). Some of the DE methods introduced above, such as edgeR and
limma, can be directly used for exon-​level DS analysis to detect differential
usage of exons. Specially designed exon-​level DS tools include DEXSeq [95]
and JunctionSeq [96]. DEXSeq, as an example, implements a test for differ-
ential exon usage between experimental conditions based on relative exon
usage, which is defined as the number of reads that map to an exon divided
by the total number of reads that map to the entire gene.
Event-​based methods focus on alternative splicing events, such as
skipped exons, alternative 3’/​5’ splicing sites, or retained introns. These
methods quantify splicing events using “percent-​spliced-​in,” or Psi (ψ),
as a measure of the frequency of an alternative splicing event into final
transcripts. Examples of event-​based methods include MISO [97], rMATS
[98], MAJIQ [99], SpliceSeq, SUPPA2 [100], SplAdder [101], Leafcutter
[102], and Whippet [103].
Isoform-​centric methods attempt to reconstruct different splicing isoforms
first, and then quantify their abundance levels in samples, prior to applying
statistical tests to detect those that show differential expression between
conditions. Previously introduced DE methods Cuffdiff 2 and EBSeq are in
this category when performing DS analysis. DiffSplice, as another example,
first maps reads to the reference genome using tools such as MapSplice, and
Transcriptomics by Bulk RNA-Seq 137

then constructs splice graph to detect alternatively spliced regions in the


form of alternative splicing modules. After estimation of their abundance,
DiffSplice uses nonparametric permutation to test for DS between experi-
mental groups.

7.4 Visualization of RNA-​Seq Data


RNA-​ seq data visualization is required to explore trends and patterns
embedded in a dataset, verify results from an established analytic workflow,
and furthermore, appreciate the complexity in gene transcription, including
alternative splicing. There are a variety of data visualization tools that can
be used for data exploration and verification of findings, including those
offered in R through packages such as ggplot2, plotly, reshape, and viridis.
Histogram, boxplot, density, violin, and volcano plots can be created using
these tools to explore a dataset, while PCA plot, heatmap, and dendrogram
can be used to provide a summarized view of the data. Verification of findings
using the same tools helps confirm that a proper workflow including data
model is used. Among the tools used to map RNA-​seq reads to the reference
genome to develop a better understanding of the complexity of gene tran-
scription, IGV and Integrated Genome Browser (IGB) are often used. In add-
ition, RNA-​seq data can be exported as custom tracks for display in a genome
browser such as the UCSC Genome Browser. For visualization of alternative
splicing, tools like SpliceSeq and JunctionSeq have their own built-​in visual-
ization capabilities. DiffSplice generates GFF-​style files that can be visualized
in the genomic browser GBrowse.

7.5 RNA-​Seq as a Discovery Tool


RNA-​seq is an important exploratory tool to study the molecular mechanisms
of biological processes. Based on the DE genes identified, new hypoth-
eses can be formulated for testing. Prior to carrying out functional studies
to establish causational relationship between change of gene expression
pattern to the biological process of interest, the first step is typically to val-
idate RNA-​seq findings with other experimental techniques, most commonly
Western blotting and quantitative PCR. Beyond DE gene detection for basic
research, the rich information in RNA-​seq data can also be used clinically to
develop new ways to classify diseases and improve diagnostic outcomes. For
example, with combined deployment of informative gene feature selection
138 Next-Generation Sequencing Data Analysis

and machine learning techniques RNA-​seq profiles are used to stratify breast
cancer into different subtypes [104]. As another example, RNA sequencing
has significantly improved diagnostic outcome for hereditary cancer by
resolving uncertainties from DNA genetic sequencing alone [105].
Besides interrogating currently catalogued genes, RNA-​ seq, being an
unbiased approach, is also a powerful technology for discovering novel
transcripts, splicing events, and other transcription-​ related phenomena.
RNA-​seq studies of the transcriptional landscape of the genome have found
that besides protein-​coding regions, the majority of the genome produces
RNA transcripts. The finding that 75% of the human genome is transcribed
(see Chapter 3), made with extensive use of RNA-​seq, shows the power of
this technology in discovering currently unknown transcripts. RNA-​seq has
also been used to discover novel alternative splicing isoforms. For example,
the discovery of circular RNAs (also see Chapter 3), which are formed as a
result of non-​canonical RNA splicing, is also due to the application of RNA-​
seq [106]. RNA-​seq has also been applied to uncover other transcription-​
related phenomena, such as gene fusion. Gene fusion is caused by genomic
rearrangement and is a common occurrence under certain conditions such as
cancer. Because RNA-​seq has the capability to locate transcripts generated
from a fused gene, detection of gene fusion events has been greatly
facilitated by this powerful technology [107, 108]. With rapid technological
developments in advancing RNA sequencing as an even more powerful dis-
covery tool, RNA-​seq has now entered into a new era –​single-cell RNA-​seq,
which is covered next.

References
1. Li J, Fu C, Speed TP, Wang W, Symmans WF. Accurate RNA sequencing from
formalin-​fixed cancer tissue To represent high-​quality transcriptome from
frozen tissue. JCO Precis Oncol 2018, 2:PO.17.00091.
2. Parekh S, Ziegenhain C, Vieth B, Enard W, Hellmann I. The impact of amplifi-
cation on differential expression analyses by RNA-​seq. Sci Rep 2016, 6:25533.
3. Zhulidov PA, Bogdanova EA, Shcheglov AS, Vagner LL, Khaspekov GL,
Kozhemyako VB, Matz MV, Meleshkevitch E, Moroz LL, Lukyanov SA et al.
Simple cDNA normalization using kamchatka crab duplex-​specific nuclease.
Nucleic Acids Res 2004, 32(3):e37.
4. Yang L, Duff MO, Graveley BR, Carmichael GG, Chen LL. Genomewide char-
acterization of non-​polyadenylated RNAs. Genome Biol 2011, 12(2):R16.
5. Busby MA, Stewart C, Miller CA, Grzeda KR, Marth GT. Scotty: a web tool
for designing RNA-​Seq experiments to measure differential gene expression.
Bioinformatics 2013, 29(5):656–​657.
6. Bi R, Liu P. Sample size calculation while controlling false discovery rate for
differential expression analysis with RNA-​ sequencing experiments. BMC
Bioinformatics 2016, 17:146.
Transcriptomics by Bulk RNA-Seq 139

7. Wu H, Wang C, Wu Z. PROPER: comprehensive power evaluation for differ-


ential expression using RNA-​seq. Bioinformatics 2015, 31(2):233–​241.
8. Zhao S, Li CI, Guo Y, Sheng Q, Shyr Y. RnaSeqSampleSize: real data based
sample size estimation for RNA sequencing. BMC Bioinformatics 2018,
19(1):191.
9. Ching T, Huang S, Garmire LX. Power analysis and sample size estimation for
RNA-​seq differential expression. RNA 2014, 20(11):1684–​1696.
10. Auer PL, Doerge RW. Statistical design and analysis of RNA sequencing data.
Genetics 2010, 185(2):405–​416.
11. Robinson DG, Storey JD. subSeq: determining appropriate sequencing depth
through efficient read subsampling. Bioinformatics 2014, 30(23):3424–​3426.
12. Grant GR, Farkas MH, Pizarro AD, Lahens NF, Schug J, Brunk BP, Stoeckert
CJ, Hogenesch JB, Pierce EA. Comparative analysis of RNA-​Seq alignment
algorithms and the RNA-​ Seq unified mapper (RUM). Bioinformatics 2011,
27(18):2518–​2528.
13. Ryan MC, Cleland J, Kim R, Wong WC, Weinstein JN. SpliceSeq: a resource
for analysis and visualization of RNA-​Seq data on alternative splicing and its
functional impacts. Bioinformatics 2012, 28(18):2385–​2387.
14. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with
RNA-​Seq. Bioinformatics 2009, 25(9):1105–​1111.
15. Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL.
TopHat2: accurate alignment of transcriptomes in the presence of insertions,
deletions and gene fusions. Genome Biol 2013, 14(4):R36.
16. Wang K, Singh D, Zeng Z, Coleman SJ, Huang Y, Savich GL, He X, Mieczkowski
P, Grimm SA, Perou CM et al. MapSplice: accurate mapping of RNA-​seq reads
for splice junction discovery. Nucleic Acids Res 2010, 38(18):e178.
17. Au KF, Jiang H, Lin L, Xing Y, Wong WH. Detection of splice junctions
from paired-​ end RNA-​ seq data by SpliceMap. Nucleic Acids Res 2010,
38(14):4570–​4578.
18. Marco-​Sola S, Sammeth M, Guigo R, Ribeca P. The GEM mapper: fast, accurate
and versatile alignment by filtration. Nat Methods 2012, 9(12):1185–​1188.
19. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P,
Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-​ seq aligner.
Bioinformatics 2013, 29(1):15–​21.
20. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low
memory requirements. Nat Methods 2015, 12(4):357–​360.
21. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-​based genome
alignment and genotyping with HISAT2 and HISAT-​genotype. Nat Biotechnol
2019, 37(8):907–​915.
22. Wu TD, Reeder J, Lawrence M, Becker G, Brauer MJ. GMAP and GSNAP
for Genomic Sequence Alignment: Enhancements to Speed, Accuracy, and
Functionality. Methods Mol Biol 2016, 1418:283–​334.
23. Krizanovic K, Echchiki A, Roux J, Sikic M. Evaluation of tools for long read
RNA-​seq splice-​aware alignment. Bioinformatics 2018, 34(5):748–​754.
24. Liu B, Liu Y, Li J, Guo H, Zang T, Wang Y. deSALT: fast and accurate long
transcriptomic read alignment with de Bruijn graph-​based index. Genome Biol
2019, 20(1):274.
25. Marić J, Sović I, Križanović K, Nagarajan N, Šikić M. Graphmap2-​splice-​aware
RNA-​seq mapper for long reads. bioRxiv 2019, doi: https://​doi.org/​10.1101/​
720​458
140 Next-Generation Sequencing Data Analysis

26. Sahlin K, Makinen V. Accurate spliced alignment of long RNA sequencing


reads. Bioinformatics 2021, 37(24):4643–​4651.
27. Graubert A, Aguet F, Ravi A, Ardlie KG, Getz G. RNA-​SeQC 2: Efficient RNA-​
seq quality control and quantification for large cohorts. Bioinformatics 2021,
37(18):3048–​3050.
28. Wang L, Wang S, Li W. RSeQC: quality control of RNA-​seq experiments.
Bioinformatics 2012, 28(16):2184–​2185.
29. Hartley SW, Mullikin JC. QoRTs: a comprehensive toolset for quality con-
trol and data processing of RNA-​Seq experiments. BMC Bioinformatics 2015,
16:224.
30. Bushmanova E, Antipov D, Lapidus A, Prjibelski AD. rnaSPAdes: a de novo
transcriptome assembler and its application to RNA-​Seq data. GigaScience
2019, 8(9):giz100.
31. Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, Adiconis
X, Fan L, Raychowdhury R, Zeng Q et al. Full-​length transcriptome assembly
from RNA-​ Seq data without a reference genome. Nat Biotechnol 2011,
29(7):644–​652.
32. Chang Z, Li G, Liu J, Zhang Y, Ashby C, Liu D, Cramer CL, Huang X. Bridger: a
new framework for de novo transcriptome assembly using RNA-​seq data.
Genome Biol 2015, 16:30.
33. Robertson G, Schein J, Chiu R, Corbett R, Field M, Jackman SD, Mungall K,
Lee S, Okada HM, Qian JQ et al. De novo assembly and analysis of RNA-​seq
data. Nat Methods 2010, 7(11):909–​912.
34. Xie Y, Wu G, Tang J, Luo R, Patterson J, Liu S, Huang W, He G, Gu S, Li S et al.
SOAPdenovo-​Trans: de novo transcriptome assembly with short RNA-​Seq
reads. Bioinformatics 2014, 30(12):1660–​1666.
35. Schulz MH, Zerbino DR, Vingron M, Birney E. Oases: robust de novo RNA-​
seq assembly across the dynamic range of expression levels. Bioinformatics
2012, 28(8):1086–​1092.
36. Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL.
StringTie enables improved reconstruction of a transcriptome from RNA-​seq
reads. Nat Biotechnol 2015, 33(3):290–​295.
37. Kovaka S, Zimin AV, Pertea GM, Razaghi R, Salzberg SL, Pertea M.
Transcriptome assembly from long-​read RNA-​seq alignments with StringTie2.
Genome Biol 2019, 20(1):278.
38. Shumate A, Wong B, Pertea G, Pertea M. Improved transcriptome assembly
using a hybrid of long and short reads with StringTie. PLoS Comput Biol 2022,
18(6):e1009730.
39. Prjibelski AD, Puglia GD, Antipov D, Bushmanova E, Giordano D, Mikheenko
A, Vitale D, Lapidus A. Extending rnaSPAdes functionality for hybrid tran-
scriptome assembly. BMC Bioinformatics 2020, 21(Suppl 12):302.
40. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose
program for assigning sequence reads to genomic features. Bioinformatics
2014, 30(7):923–​930.
41. Anders S, Pyl PT, Huber W. HTSeq—​a Python framework to work with high-​
throughput sequencing data. Bioinformatics 2015, 31(2):166–​169.
42. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-​Seq data
with or without a reference genome. BMC Bioinformatics 2011, 12:323.
Transcriptomics by Bulk RNA-Seq 141

43. Roberts A, Pachter L. Streaming fragment assignment for real-​time analysis of


sequencing experiments. Nat Methods 2013, 10(1):71–​73.
44. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H,
Salzberg SL, Rinn JL, Pachter L. Differential gene and transcript expression
analysis of RNA-​seq experiments with TopHat and Cufflinks. Nat Protoc 2012,
7(3):562–​578.
45. Robert C, Watson M. Errors in RNA-​Seq quantification affect genes of rele-
vance to human disease. Genome Biol 2015, 16:177.
46. Bray NL, Pimentel H, Melsted P, Pachter L. Near-​optimal probabilistic RNA-​
seq quantification. Nat Biotechnol 2016, 34(5):525–​527.
47. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast
and bias-​ aware quantification of transcript expression. Nat Methods 2017,
14(4):417–​419.
48. Patro R, Mount SM, Kingsford C. Sailfish enables alignment-​free isoform quan-
tification from RNA-​seq reads using lightweight algorithms. Nat Biotechnol
2014, 32(5):462–​464.
49. Wu DC, Yao J, Ho KS, Lambowitz AM, Wilke CO. Limitations of alignment-​
free tools in total RNA-​seq quantification. BMC Genomics 2018, 19(1):510.
50. Bullard JH, Purdom E, Hansen KD, Dudoit S. Evaluation of statistical methods
for normalization and differential expression in mRNA-​ Seq experiments.
BMC Bioinformatics 2010, 11:94.
51. Hansen KD, Irizarry RA, Wu Z. Removing technical variability in RNA-​seq
data using conditional quantile normalization. Biostatistics 2012, 13(2):204–​216.
52. Love MI, Huber W, Anders S. Moderated estimation of fold change and dis-
persion for RNA-​seq data with DESeq2. Genome Biol 2014, 15(12):550.
53. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for
differential expression analysis of digital gene expression data. Bioinformatics
2010, 26(1):139–​140.
54. Robinson MD, Oshlack A. A scaling normalization method for differential
expression analysis of RNA-​seq data. Genome Biol 2010, 11(3):R25.
55. Kadota K, Nishiyama T, Shimizu K. A normalization strategy for comparing
tag count data. Algorithms Mol Biol 2012, 7(1):5.
56. Sun J, Nishiyama T, Shimizu K, Kadota K. TCC: an R package for comparing
tag count data with robust normalization strategies. BMC Bioinformatics 2013,
14 :219.
57. Li J, Witten DM, Johnstone IM, Tibshirani R. Normalization, testing, and
false discovery rate estimation for RNA-​sequencing data. Biostatistics 2012,
13(3):523–​538.
58. Manimaran S, Selby HM, Okrah K, Ruberman C, Leek JT, Quackenbush J,
Haibe-​Kains B, Bravo HC, Johnson WE. BatchQC: interactive software for
evaluating sample and batch effects in genomic data. Bioinformatics 2016,
32(24):3836–​3838.
59. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expres-
sion data using empirical Bayes methods. Biostatistics 2007, 8(1):118–​127.
60. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-​seq data using
factor analysis of control genes or samples. Nat Biotechnol 2014, 32(9):896–​902.
61. Leek JT. svaseq: removing batch effects and other unwanted noise from
sequencing data. Nucleic Acids Res 2014, 42(21):e161.
142 Next-Generation Sequencing Data Analysis

62. Zhang Y, Parmigiani G, Johnson WE. ComBat-​seq: batch effect adjustment for
RNA-​seq count data. NAR Genom Bioinform 2020, 2(3):lqaa078.
63. Marioni JC, Mason CE, Mane SM, Stephens M, Gilad Y. RNA-​ seq: an
assessment of technical reproducibility and comparison with gene expression
arrays. Genome Res 2008, 18(9):1509–​1517.
64. Anders S, Huber W. Differential expression analysis for sequence count data.
Genome Biol 2010, 11(10):R106.
65. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers
differential expression analyses for RNA-​sequencing and microarray studies.
Nucleic Acids Res 2015, 43(7):e47.
66. Frazee AC, Pertea G, Jaffe AE, Langmead B, Salzberg SL, Leek JT. Ballgown
bridges the gap between transcriptome assembly and expression analysis. Nat
Biotechnol 2015, 33(3):243–​246.
67. Hardcastle TJ, Kelly KA. baySeq: empirical Bayesian methods for identi-
fying differential expression in sequence count data. BMC Bioinformatics
2010, 11:422.
68. Trapnell C, Hendrickson DG, Sauvageau M, Goff L, Rinn JL, Pachter L.
Differential analysis of gene regulation at transcript resolution with RNA-​seq.
Nat Biotechnol 2013, 31(1):46–​53.
69. Wang L, Feng Z, Wang X, Wang X, Zhang X. DEGseq: an R package for iden-
tifying differentially expressed genes from RNA-​seq data. Bioinformatics 2010,
26(1):136–​138.
70. Leng N, Dawson JA, Thomson JA, Ruotti V, Rissman AI, Smits BM, Haag
JD, Gould MN, Stewart RM, Kendziorski C. EBSeq: an empirical Bayes hier-
archical model for inference in RNA-​seq experiments. Bioinformatics 2013,
29(8):1035–​1043.
71. Li J, Tibshirani R. Finding consistent patterns: a nonparametric approach for
identifying differential expression in RNA-​Seq data. Stat Methods Med Res
2013, 22(5):519–​536.
72. Tarazona S, Furio-​Tari P, Turra D, Pietro AD, Nueda MJ, Ferrer A, Conesa
A. Data quality aware analysis of differential expression in RNA-​seq with
NOISeq R/​Bioc package. Nucleic Acids Res 2015, 43(21):e140.
73. Corchete LA, Rojas EA, Alonso-​ Lopez D, De Las Rivas J, Gutierrez NC,
Burguillo FJ. Systematic comparison and assessment of RNA-​seq procedures
for gene expression quantitative analysis. Sci Rep 2020, 10(1):19737.
74. Stupnikov A, McInerney CE, Savage KI, McIntosh SA, Emmert-​ Streib F,
Kennedy R, Salto-​Tellez M, Prise KM, McArt DG. Robustness of differen-
tial gene expression analysis of RNA-​seq. Comput Struct Biotechnol J 2021,
19:3470–​3481.
75. Feng J, Meyer CA, Wang Q, Liu JS, Shirley Liu X, Zhang Y. GFOLD: a
generalized fold change for ranking differentially expressed genes from RNA-​
seq data. Bioinformatics 2012, 28(21):2782–​2788.
76. Claverie JM, Ta TN. ACDtool: a web-​server for the generic analysis of large
data sets of counts. Bioinformatics 2019, 35(1):170–​171.
77. Audic S, Claverie JM. The significance of digital gene expression profiles.
Genome Res 1997, 7(10):986–​995.
78. Benjamini Y, Hochberg Y. Controlling the false discovery rate –​a practical
and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 1995,
57(1):289–​300.
Transcriptomics by Bulk RNA-Seq 143

79. Gene Ontology C. The Gene Ontology resource: enriching a GOld mine.
Nucleic Acids Res 2021, 49(D1):D325–​D334.
80. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new
perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res
2017, 45(D1):D353–​D361.
81. Rodchenkov I, Babur O, Luna A, Aksoy BA, Wong JV, Fong D, Franz M,
Siper MC, Cheung M, Wrana M et al. Pathway Commons 2019 update: inte-
gration, analysis and exploration of pathway data. Nucleic Acids Res 2020,
48(D1):D489–​D497.
82. Martens M, Ammar A, Riutta A, Waagmeester A, Slenter DN, Hanspers K, R
AM, Digles D, Lopes EN, Ehrhart F et al. WikiPathways: connecting commu-
nities. Nucleic Acids Res 2021, 49(D1):D613–​D621.
83. Gillespie M, Jassal B, Stephan R, Milacic M, Rothfels K, Senff-​Ribeiro A, Griss
J, Sevilla C, Matthews L, Gong C et al. The reactome pathway knowledgebase
2022. Nucleic Acids Res 2022, 50(D1):D687–​D692.
84. Xie Z, Bailey A, Kuleshov MV, Clarke DJB, Evangelista JE, Jenkins SL,
Lachmann A, Wojciechowicz ML, Kropiwnicki E, Jagodnik KM et al. Gene set
knowledge discovery with Enrichr. Curr Protoc 2021, 1(3):e90.
85. Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for
RNA-​seq: accounting for selection bias. Genome Biol 2010, 11(2):R14.
86. Eden E, Navon R, Steinfeld I, Lipson D, Yakhini Z. GOrilla: a tool for discovery
and visualization of enriched GO terms in ranked gene lists. BMC bioinfor-
matics 2009, 10:48.
87. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, Vilo J.
g:Profiler: a web server for functional enrichment analysis and conversions of
gene lists (2019 update). Nucleic Acids Res 2019, 47(W1):W191–​W198.
88. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis
of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009,
4(1):44–​57.
89. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrich-
ment analysis and candidate gene prioritization. Nucleic Acids Res 2009,
37(Web Server issue):W305–​311.
90. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA,
Paulovich A, Pomeroy SL, Golub TR, Lander ES et al. Gene set enrichment
analysis: a knowledge-​based approach for interpreting genome-​wide expres-
sion profiles. Proc Natl Acad Sci U S A 2005, 102(43):15545–​15550.
91. Doncheva NT, Morris JH, Gorodkin J, Jensen LJ. Cytoscape stringapp:
network analysis and visualization of proteomics data. J Proteome Res 2019,
18(2):623–​632.
92. Montojo J, Zuberi K, Rodriguez H, Kazi F, Wright G, Donaldson SL, Morris Q,
Bader GD. GeneMANIA Cytoscape plugin: fast gene function predictions on
the desktop. Bioinformatics 2010, 26(22):2927–​2928.
93. Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A,
Fridman WH, Pages F, Trajanoski Z, Galon J. ClueGO: a Cytoscape plug-​in
to decipher functionally grouped gene ontology and pathway annotation
networks. Bioinformatics 2009, 25(8):1091–​1093.
94. Merico D, Isserlin R, Stueker O, Emili A, Bader GD. Enrichment map: a
network-​based method for gene-​set enrichment visualization and interpret-
ation. PLoS One 2010, 5(11):e13984.
144 Next-Generation Sequencing Data Analysis

95. Anders S, Reyes A, Huber W. Detecting differential usage of exons from


RNA-​seq data. Genome Res 2012, 22(10):2008–​2017.
96. Hartley SW, Mullikin JC. Detection and visualization of differential splicing
in RNA-​Seq data with JunctionSeq. Nucleic Acids Res 2016, 44(15):e127.
97. Katz Y, Wang ET, Airoldi EM, Burge CB. Analysis and design of RNA
sequencing experiments for identifying isoform regulation. Nat Methods
2010, 7(12):1009–​1015.
98. Shen S, Park JW, Lu ZX, Lin L, Henry MD, Wu YN, Zhou Q, Xing Y. rMATS:
Robust and flexible detection of differential alternative splicing from repli-
cate RNA-​Seq data. Proc Natl Acad Sci U S A 2014, 111(51):E5593–​E5601.
99. Vaquero-​Garcia J, Barrera A, Gazzara MR, Gonzalez-​Vallinas J, Lahens NF,
Hogenesch JB, Lynch KW, Barash Y. A new view of transcriptome com-
plexity and regulation through the lens of local splicing variations. Elife 2016,
5:e11752.
100. Trincado JL, Entizne JC, Hysenaj G, Singh B, Skalic M, Elliott DJ, Eyras E.
SUPPA2: fast, accurate, and uncertainty-​aware differential splicing analysis
across multiple conditions. Genome Biol 2018, 19(1):40.
101. Kahles A, Ong CS, Zhong Y, Ratsch G. SplAdder: identification, quan-
tification and testing of alternative splicing events from RNA-​Seq data.
Bioinformatics 2016, 32(12):1840–​1847.
102. Li YI, Knowles DA, Humphrey J, Barbeira AN, Dickinson SP, Im HK, Pritchard
JK. Annotation-​free quantification of RNA splicing using LeafCutter. Nat
Genet 2018, 50(1):151–​158.
103. Sterne-​Weiler T, Weatheritt RJ, Best AJ, Ha KCH, Blencowe BJ. Efficient
and Accurate Quantitative Profiling of Alternative Splicing Patterns of Any
Complexity on a Laptop. Mol Cell 2018, 72(1):187–​200 e186.
104. Cascianelli S, Molineris I, Isella C, Masseroli M, Medico E. Machine learning
for RNA sequencing-​based intrinsic subtyping of breast cancer. Sci Rep 2020,
10(1):14071.
105. Karam R, Conner B, LaDuca H, McGoldrick K, Krempely K, Richardson
ME, Zimmermann H, Gutierrez S, Reineke P, Hoang L et al. Assessment of
Diagnostic Outcomes of RNA Genetic Testing for Hereditary Cancer. JAMA
Netw Open 2019, 2(10):e1913900.
106. Salzman J, Gawad C, Wang PL, Lacayo N, Brown PO. Circular RNAs are the
predominant transcript isoform from hundreds of human genes in diverse
cell types. PLoS One 2012, 7(2):e30733.
107. Davare MA, Tognon CE. Detecting and targeting oncogenic fusion proteins
in the genomic era. Biol Cell 2015, 107(5):111–​129.
108. Haas BJ, Dobin A, Li B, Stransky N, Pochet N, Regev A. Accuracy assessment
of fusion transcript detection via read-​mapping and de novo fusion tran-
script assembly-​based methods. Genome Biol 2019, 20(1):213.
8 
Transcriptomics by Single-Cell RNA-​Seq

Bulk RNA-​seq, as covered in Chapter 7, reveals the transcriptome at the


level of an organ, a tissue, or a population of primary or cultured cells. Bulk
RNA-​seq is very effective to reveal overall gene expression change under
contrasting conditions, but the detected change is a result of comparing
average gene expression of all cells between conditions. It was highly chal-
lenging, if not impossible, to analyze the transcriptome of each cell individu-
ally not long ago. But with the recent rapid technological developments in
the single-cell sequencing field, interrogating the transcriptome of thousands
to tens of thousands of single cells simultaneously has become a reality.
For the first time single-cell RNA-​seq, or scRNA-​seq, enables researchers to
study cell-​to-​cell variation, even among those that appear to be similar mor-
phologically and reside in close proximity. Single-​cell RNA-​seq has offered
unprecedented opportunities for the biomedical community to identify dis-
tinct subpopulations of cells in a heterogenous population, characterize their
functional states, and infer cellular trajectories during differentiation.
While creating unprecedented opportunities, scRNA-​ seq also presents
new challenges that do not exist in bulk RNA-​seq. Some of the challenges
come from the tremendous amount of cellular heterogeneity in a typical
tissue sample, and therefore the richness of bio-​information embedded in the
scRNA-​seq dataset from the much-​increased resolution and change of scale.
Other challenges, however, are due to the inherent characteristics of scRNA-​
seq data. Compared to bulk RNA-​seq data, scRNA-​seq data is sparser, i.e.,
containing a lot of zeros (often called dropouts), and has higher technical
noise. This signal sparsity is partially biological as a result of normal gene
transcription oscillation at the single-cell level. In the meantime, this is also
caused by technical challenges. Based on estimation, a typical mammalian
cell has on average 360,000 mRNA molecules representing 12,000 different
transcripts, with wide variation from cell type to cell type [1]. By weight,
a cell typically contains ~10 pg RNA, of which mRNA molecules are only
a small fraction (1–​5%) [2]. Generating scRNA-​seq signals from such low
quantities of cellular mRNA is technically challenging. For example, the
rate of mRNA molecular capture is still quite low in the range of 10–​20%
[3–​5]. Such technical challenges, along with non-​exhaustive sequencing of
captured molecules, lead to signal stochasticity, false-​negative detection, and

DOI: 10.1201/9780429329180-11 145


146 Next-Generation Sequencing Data Analysis

high measurement noise in the scRNA-​seq data. To effectively extract the rich
information embedded in the scRNA-​seq data, these characteristics demand
development and deployment of algorithms and tools that are different from
those designed for bulk RNA-​seq.
Furthermore, the questions that can be answered from scRNA-​seq data are
beyond those from bulk RNA-​seq. Detection of cell-​to-​cell variation, identifi-
cation and visualization of different cell types/​identities in a population, and
inference of cellular developmental trajectories are all new realms that have
emerged from the development of scRNA-​seq. Some other data analytical
topics such as data preprocessing, normalization, batch effect correction, and
clustering are also significantly different from those covered in Chapter 7,
and are presented in detail in this chapter. On topics that are similar to and/​
or have significant overlap with those covered in Chapter 7, such as identifi-
cation and functional analysis of differentially expressed genes, only aspects
that are specific to or need adjustment for scRNA-​seq data are presented to
avoid redundancy.

8.1 Experimental Design
8.1.1 Single-Cell RNA-​Seq General Approaches
The landscape of single-​cell sequencing, including RNA-​seq, has been rap-
idly evolving. There have been a multitude of strategies or platforms in
existence today, while new ones are continuously being developed and
existing ones improved upon. In general, existing scRNA-​ seq strategies
can be divided into two broad approaches, i.e., low-​and high-​throughput,
based on the number of cells that can be analyzed simultaneously at a time.
Low-​throughput methods can process up to a few hundred cells at a time,
while those of the high-​throughput approach allow simultaneous analysis
of thousands to tens of thousands of cells, or even more with the technolo-
gies continuing to evolve. A good example of the low-​throughput approach
is Smart-​seq3 [6]. The low-​throughput approach suits situations where the
focus is on a small number of cells that need detailed molecular character-
ization. This approach has higher sensitivity, which leads to the identifica-
tion of more transcripts and genes from each cell. It also provides full-​length
coverage of the transcriptome, thus enabling recognition of different splicing
isoforms and allelic gene expression.
There are currently a multitude of platforms for the high-​ throughput
approach. These platforms include Drop-​seq [7], inDrop [8], sci-​RNA-​seq for
single-cell combinatorial indexing RNA sequencing [9], and 10× Chromium
[10]. Among these various platforms, the commercially available 10×
Transcriptomics by Single-Cell RNA-Seq 147

Chromium platform has been more widely adopted so far in the biomed-
ical research community. Benchmark studies have shown that 10× generally
has more consistent performance, better sensitivity and precision, and lower
technical noise [11, 12]. In general, the high-​throughput approach is more
suitable to study cellular heterogeneity in a large population of cells. The
transcript detection sensitivity is typically lower with this approach, as a
result the data is sparser. The detection and counting of transcripts are usu-
ally based on sequencing of either the 3’ or 5’ end and not full length. Because
of such differences between the two approaches, it is advisable to evaluate
the particular needs of a project in order to decide which approach is more
appropriate.
Despite the differences, the two general approaches share basically the
same workflow in wet lab operational procedures. As detailed in Section
8.2, for both approaches single cells need to be prepared from dissociation of
input materials, such as an organ or a tissue biopsy, followed by partitioning
of the cells. The mechanism of cell partitioning varies with the approach
and particular platform chosen, e.g., cells can be partitioned into 96-​or 384-​
well plates (such as for the low-​throughput approach), microfluidic droplets
(used by many of the high-​throughput platforms), etc. On the 10× Chromium
platform, single cells are partitioned into nanoliter-​scale GEMs (or Gel beads-​
in-​EMulsion), with each GEM carrying a unique barcode. After partitioning
cells are lysed to release cellular RNA, which is then reverse transcribed into
cDNA. This is followed by cDNA amplification and subsequently library
construction. Because of the shared commonalities in lab operation as well as
data analysis, the two general approaches are not covered separately in the
following sections unless otherwise noted.

8.1.2 Cell Number and Sequencing Depth


As many scRNA-​seq projects aim to investigate cellular heterogeneity in a
large cell population, to adopt the high-​throughput approach some of the
major technical questions to address first include how many cells to sequence,
how deep to sequence, and how to balance cell number and sequencing depth
in cases of budgetary constraints to maximize the amount of information to
be obtained. On the question of how many cells to sequence, this depends on
(1) the number of cell types present in the target population, (2) the minimum
fraction of any of the cell types, and (3) the minimum number of cells desired
to be observed in any of the cell types. Based on this information, negative
binomial (NB) distribution can be used to estimate the number of cells to
sample. For example, if 10 cell types are expected in a population, with each
type being present at a minimum fraction of 3% in the total population, in
order to observe at least 20 cells in any of the cell types, we will need to sample
at least 1,104 cells in total at the 95% confidence level [13]. If the needed infor-
mation on the heterogeneity of the population is not available, it is advisable
to first conduct a pilot study using the maximum number of cells possible.
148 Next-Generation Sequencing Data Analysis

The maximum allowable cell number varies with platform, for example, on
the current configuration of 10× Chromium 10,000 cells is the upper limit. For
any droplet-​based platforms including the Chromium, the limit on maximum
cell number is affected by the law of Poisson distribution, as the loading of
single cells into droplets (or GEMs for Chromium) is a Poisson process. This
process leads to the formation of doublets (or multiplets), i.e., two (or more)
cells being loaded into the same droplet/​GEM and treated as one cell, and
the rate of their formation follows Poisson statistics. To ensure scRNA-​seq
data quality, the rate of doublets/​multiplets needs to be controlled at a man-
ageable level (usually <5%).
On the question of sequencing depth, this depends on how transcription-
ally active cells are in the sample, and the diversity of their transcriptomes.
Without this knowledge a priori, the generally suggested depth for 3’ (or
5’) end gene expression profiling on the 10× Chromium platform is a min-
imum of 20,000 reads per cell using the current v3 chemistry. The number of
detected genes at this depth varies with cell type, e.g., 1,000–​2,000 for periph-
eral blood mononuclear cells and 2,000–​3,000 for neurons. Sequencing depth
may be fine-​tuned based on specific project needs, and increasing sequen-
cing depth from the suggested minimum depth generally leads to identifica-
tion of more genes. But beyond a certain point (varies with cell type), further
increase in sequencing depth leads to diminished return.
On the question of how to balance cell number and sequencing depth
to maximize the amount of information obtained, the options are either
sequencing fewer cells at greater depth, or more cells with fewer reads per
cell. The former option allows identification of more transcripts and genes,
and as a result generates a more accurate picture of each cell’s transcription
status. However, the fewer cells used may not offer a sufficient representa-
tion of the cellular population under study. The latter option, on the other
hand, allows analysis of more cells to increase cell representation, but at the
expense of identifying less transcripts and genes per cell. To quantify the
tradeoff between sequencing depth and cell number, computational simula-
tion using a multivariate generative model showed that increasing sequen-
cing depth is better than increasing cell number before reaching the depth of
15,000 reads per cell, beyond which point there is a diminished return [14].
Another modeling demonstrated that under a fixed budget the strategy is
to sequence as many cells as possible at the depth of one read per gene per
cell [15]. Commonly used scRNA-​seq pipeline tools like the open source 10×
Cell Ranger [10] and zUMIs [16] have downsampling function to help deter-
mine whether the library was sequenced to saturation, or whether additional
sequencing would increase the number of detected genes cost effectively.
To further compare the two options, another factor to consider is that
many genes are often co-​regulated in a cell, forming functional modules. The
modularity of the cellular gene transcriptional system is manifested in the
extensive gene-​gene covariance embedded in the sequencing data, which
Transcriptomics by Single-Cell RNA-Seq 149

suggests that scRNA-​seq data is inherently low dimensional [17]. Because


of this nature, a number of downstream data analysis steps are quite robust
and can tolerate technical noise caused by shallow sequencing. Because of
this, for many studies especially those that involve highly heterogeneous
cell populations, limited resources are better spent on sequencing more cells
instead of increasing sequencing depth. For studies in which cells are more
homogeneous with more nuanced differences, sequencing fewer cells at
greater depth might offer better distinguishing power. In this situation, the
depth of 1–​2 million reads per cell has been shown to be sufficient, beyond
which detection sensitivity barely increases [18, 19].

8.1.3 Batch Effects Minimization and Sample Replication


The general principles of factorial design, randomization, and replication
as covered in the last chapter for bulk RNA-​seq equally apply to single-
cell studies. In an ideal design, samples are prepared and sequenced sim-
ultaneously, with different groups or experimental conditions randomly
distributed across all samples. For sequencing, all libraries should be mixed
and sequenced together across all lanes. Compared to bulk RNA-​seq, how-
ever, scRNA-​seq samples are more often divided into batches. This is usually
due to practical considerations, such as technical (as it is more challenging
to prepare and process a large number of single-​cell samples at once), or
budgetary (a small-​scale pilot run is often run first due to the associated high
cost of running scRNA-​seq). To minimize batch effects, the same principles
should still be adhered to as much as possible within the same batch. For
example, to study lung cell compositional change before and after an infec-
tion, when samples are divided into batches, within each batch dissociated
cells should be prepared from lung tissue dissected from infected and con-
trol animals of the same age, with the procedure performed by a single
operator following the same procedure. For subsequent cell partitioning,
the cells should be partitioned using the same microfluidic chip if using
10× Chromium. If multiple chips are needed, loading of samples to chips
and wells should be randomized and balanced. For sequencing, libraries
prepared from all samples need to be barcoded differently, mixed together,
and loaded onto multiple lanes. An unbalanced design, such as processing
and sequencing of infected and control samples on two separate chips and
different sequencer lanes, can make it challenging to distinguish cell compos-
itional change caused by the infection from the variation resulted from the
technical differences in processing and sequencing.
While it is a common practice to use biological replicates in bulk RNA-​seq
to measure within-​group variation, there is still a lack of consensus on the use
of replicates in scRNA-​seq experiments. This is partly because scRNA-​seq
data contains more information than bulk RNA-​seq, and therefore there are
more diverse analyses that can be performed on scRNA-​seq data beyond the
150 Next-Generation Sequencing Data Analysis

identification of differentially expressed genes. How biological replicates can


be incorporated into and help these analyses is still not very clear. As a gen-
eral trend, use of biological replicates has become more common, allowing
examination of reproducibility on these analyses. The number of biological
replicates to run can vary based on the specific questions asked, as well as
the computational power available to the researcher as inclusion of biological
replicates can significantly increase the demand on computing resources in
some of the analytic steps. It is not necessary to run technical replicates if
using a standardized platform such as 10× Chromium.

8.2 Single-Cell Preparation, Library Construction,


and Sequencing
8.2.1 Single-Cell Preparation
Generating high-​ quality scRNA-​ seq data is a multi-​ step process (see
Figure 8.1), and preparing high-​quality single cells is an important first step.
To prepare the large number of cells needed for high-​throughput scRNA-​seq,
cell preparatory procedure needs to be optimized for each target tissue in order
to generate cells that are of high viability, with minimal cell death/​debris and
no cell aggregation. If present, dead and dying cells need to be removed first
before proceeding. If they are not removed, the RNA they release to the envir-
onment from their breakdown leads to high levels of ambient RNA, which in
turn results in increased background noise [20] that is difficult, if not impos-
sible, to remove bioinformatically during data analysis. Preparing cells from
liquid tissues such as blood and bone marrow is relatively straightforward.
When starting from solid tissues, enzymatic and mechanical dissociation is
required first to break down extracellular matrix and cleave cell-​cell junctions.
If certain types of cells need to be enriched, approaches such as fluorescence-​
activated cell sorting (FACS), microchip-​, or magnetic-​activated cell sorting
are commonly used. If using FACS, gentle settings such as large nozzle sizes
and/​or slower flow velocities are recommended to minimize cell damage
and death. If using a low-​throughput platform, besides the above methods,
manual microscope-​guided precision cell picking techniques, such as micro-​
pipetting used in micromanipulation or laser capture microdissection, are
also often used. If using these techniques, target cells can be directly deposited
in lysis buffer that contains RNase inhibitor.
Cell preparatory procedures may also lead to alteration of cellular gene
expression profiles. For example, early stress response genes such as Fos and
Jun have been found to be expressed after cell dissociation and FACS [21].
To avoid such unintended changes, once prepared single cells should be
processed as quickly as possible without delay to subsequent steps, including
newgenrtpdf
Transcriptomics by Single-Cell RNA-Seq
Cell Barcode incorporated

Preprocessing Cell 1 Cell 2 Cell 3 . . . Cell N

Single Cell Lysis Combining & Generation of ...


Gene 1 220 23 1 150
Cell cDNA Synthesis Libraries & Gene-Cell
Partitioning Library Prep Sequencing Matrix Gene 2 0 3 0 ... 1
Gene 3 2 13 5 ... 0

...
...

...
...
...
...

Gene M 18 12 0 ... 0

FIGURE 8.1
Single-cell RNA-​seq general lab process. Single cells are first partitioned into individual droplets, wells, tubes, etc. Once partitioned, cells are lysed to
release RNA for reverse transcription into cDNA. During cDNA synthesis and subsequent sequencing library preparation, cell-​specific barcodes are
incorporated to track transcripts from each cell. After sequencing of scRNA-​seq libraries, reads are demultiplexed as part of preprocessing to reveal
transcripts from each cell using cell barcodes. Counting of transcripts from each cell generates a gene-​cell matrix for further analyses.

151
152 Next-Generation Sequencing Data Analysis

cell partitioning, lysis, and cDNA conversion. If immediate processing is not


possible due to practical limitations (e.g., distance, time, and processing cap-
acity), alternative strategies including cryopreservation and fixation can be
used. For cryopreservation, dimethyl sulfoxide (DMSO) is often added to
prevent the formation of ice crystals that result in cell damage. If fixation is
preferred, methanol can be used. Among such methods it has been found
that DMSO cryopreservation offers a better approach than fixation, or other
specially developed preservatives [22].

8.2.2 Single Nuclei Preparation


Variations in cell size and shape may prevent conduct of scRNA-​seq on cer-
tain platforms. For example, the width of the microfluidic channel used in
the 10× Chromium controller is 50–​60 μm. Most cells in the body are below
this size in the range of 10–​15 μm in diameter. Some cells, however, are larger
(e.g., human egg cells, 100 μm in diameter), or have highly sophisticated
shape (e.g., neurons, with dendrites and axons that can reach over one meter
in length), preventing their passing through the channel. One solution for
such cells is to use single nucleus RNA-​seq (or snRNA-​seq). Side-​by-​side
comparisons of scRNA-​seq and snRNA-​seq data have shown that they reveal
similar transcriptional profiles [23–​25], lending support to the use of snRNA-​
seq as a surrogate for scRNA-​ seq. These comparative studies have also
shown that snRNA-​seq data contain more reads that map to intronic regions,
and the number of genes detected from snRNA-​seq is in general lower than
that from scRNA-​seq. These observations are consistent with the fact that the
population of transcripts assayed by snRNA-​seq is the nuclear, still being​
processed portion of the cell’s entire transcriptome (nuclear and cytoplasmic)
assayed by scRNA-​seq. As detailed in Chapter 3, to generate mature mRNA,
the spliceosome needs to remove introns from pre-​mRNAs before mature
mRNAs are exported out of the nucleus to the cytoplasm.
Besides accommodating cells of unusual sizes and shapes, snRNA-​seq
may also offer advantages over scRNA-​seq in sample prep and availability
because of procedural differences between the two approaches. For example,
cells in certain tissues (such as the retina) are more prone to damage and cell
death during tissue dissociation, which can lead to their underrepresentation
in the final scRNA-​seq data. In processing such tissues snRNA-​seq can offer
an advantage because single nuclei prep is usually faster, and it is usually
easier to maintain the intactness of prepared nuclei than entire cells [26]. With
snRNA-​seq, archival frozen tissues may also be used [25], which as indicated
in the last section is an issue with scRNA-​seq if appropriate cryopreservation
processes were not followed for freezing the tissues. In addition, for cells that
have a high tendency to aggregate after dissociation, such as monocytes and
granulocytes, snRNA-​seq also offers a good alternative since prepared single
nuclei have a low propensity to aggregate.
Transcriptomics by Single-Cell RNA-Seq 153

8.2.3 Library Construction and Sequencing


To construct scRNA/​snRNA-​seq libraries, the released RNA from lysed cells
(or nuclei) is first reverse transcribed into cDNA. This step is most commonly
carried out using oligo-​dT primers that target polyadenylated transcripts. To
generate full-​length cDNA, the so-​called SMART technology, or Switching
Mechanism at 5’ End of RNA Template [27], is often used. Because of the
low amounts of cDNA generated from each cell, PCR amplification is needed
to boost cDNA quantity. To make the amplified cDNA into sequenceable
libraries, different scRNA-​ seq platforms use different strategies. For the
10× Chromium, the amplified cDNA is enzymatically fragmented into
smaller pieces to accommodate Illumina shorts read sequencing. The cDNA
fragments are then end repaired, followed by A-​ tailing and ligation of
Illumina adapters. A subsequent PCR is the last step of library construction
that incorporates sample barcodes, as well as the rest of Illumina adapter
sequences required for sequencing. For the low-​ throughput Smart-​ seq3,
a quicker, Nextera-​based method is used, in which the amplified cDNA is
fragmented using tagmentation, followed by a library amplification process
prior to sequencing.
As a result of cDNA amplification, one original transcript molecule
generates multiple copies and therefore is represented multiple times in
the final sequencing library. Without correcting for this amplification effect,
reads derived from PCR duplicates of the same original transcript cannot be
differentiated from those from different mRNA molecules transcribed from
the same gene (the true detection target). To correct for this effect and thereby
reduce experimental noise, unique molecular identifiers (or UMIs) are used,
which are essentially a large number of randomly synthesized, unique
nucleotide combinations. UMIs are introduced into each cDNA before the
amplification step, and after amplification all PCR products from the same
original cDNA will carry the same UMI. After sequencing, reads carrying
the same UMI are bioinformatically collapsed into one, thereby removing
the amplification effect. The use of UMI has been adopted by both high-​and
low-​throughput scRNA-​seq approaches.
The specifics of sequencing scRNA-​seq libraries may vary depending on
the particular platform chosen. To sequence 10× libraries, for example, the
specific read length required of Illumina sequencing for 3’ v3.1 dual indexed
libraries (Figure 8.2) is: Read 1–​28 bp, covering cell barcode (16 bp) and UMI
(12 bp); Index 1 (i7) –​10 bp; Index 2 (i5) –​10 bp; Read 2–​90 bp, covering
cDNA insert. For Smart-​seq3 libraries, regular single end 50 or 75 bp sequen-
cing can be used for general gene expression profiling, and paired end 150
bp sequencing can be used for splicing isoform and/​or allelic expression
analyses. With regard to sequencing depth, the reader should refer to above,
Section 8.1.2.
154 Next-Generation Sequencing Data Analysis

Read 1:28
Sample 10x BC +UMI Sample
Index (i5:10) Index (i7:10)

P5 TruSeq 10x UMI Poly(dT)VN TruSeq P7


Read 1 Barcode Read 2:90 Read 2
Insert

FIGURE 8.2
Structure of a 10× scRNA-​seq 3’ library. To sequence such a library using Illumina sequencers,
Read 1 covers 10× barcodes (16 bp) that track individual cells and UMI (12 bp) for removal of
PCR duplicates, while Read 2 is used to sequence actual cDNA fragments for gene detection.
The dual sample indices (i5 and i7, 10 bp each) are used to separate reads into different samples,
each of which may contain thousands of cells. (This illustration is based on 10× scRNA-​seq v3.1
chemistry. Image provided by 10× Genomics.)

8.3 Preprocessing of scRNA-​Seq Data


After sequencing, raw scRNA-​seq reads need to be preprocessed before fur-
ther analysis can be conducted (Figure 8.3 provides an overview of the basic
scRNA-​seq data analysis workflow). The tasks of the preprocessing step
include assigning reads to their cells of origin, collapsing reads according to
their UMIs, quantifying the abundance of transcripts in each cell, and gener-
ating a cell-​transcript count matrix, among others to be detailed next. It should
be noted that these tasks put high demands on computational resources, as
the substantial increase in resolution to the single-cell level is associated with
significant increase in raw data volume in comparison to bulk RNA-​seq. For
preprocessing of 10× scRNA-​seq data using the Cell Ranger software, for
example, the recommended requirements are a Linux system with 16 CPU
cores, 128 GB RAM, and 1 TB free disk space.

8.3.1 Initial Data Preprocessing and Quality Control


As performed for other NGS applications, the original sequencing data in
the BCL format needs to be first demultiplexed (if two or more samples are
sequenced together), and converted to FASTQ files. While this is typically
achieved using tools such as Illumina’s bcl2fastq, if using the 10× Chromium
platform, the 10× Cell Ranger, which offers a series of pipelines for ana-
lyzing 10× scRNA-​seq data, has a built-​in bcl2fastq wrapper in its “cellranger
mkfastq” pipeline. For quality control (QC) of the FASTQ data, while general
NGS data QC tools such as FastQC can be used, specialized scRNA-​seq soft-
ware such as Cell Ranger provides specific scRNA-​seq QC metrics, including
total number of reads from each sample, % of Read 1 and 2 bases ≥Q30, %
of cell barcode bases ≥Q30, % of GEM barcodes from the 10× GEM barcode
“whitelist,” etc. If using the low-​throughput scRNA-​seq approach, tools such
as zUMIs can take FASTQ files as input to perform data QC. As part of initial
Transcriptomics by Single-Cell RNA-Seq 155

Demultiplexed Raw Reads

Read Alignment
& Gene Counting

Data Cleanup Post Alignment

Data Preprocessing
Normalization

Batch Effects Correction


& Data Integration

Signal Imputation

Feature Selection

Dimension Reduction
Dimension Reduction
& Visualization

Visualization

Cell Clustering
Downstream
Analyses
Cell Identity Annotation

Differential Compositional Trajectory


Expression Analysis Inference

FIGURE 8.3
Basic scRNA-​seq data analysis workflow.
156 Next-Generation Sequencing Data Analysis

data QC and preprocessing, non-​transcript sequences such as polyA and


template switching oligo (required for the SMART-​based cDNA synthesis)
need to be checked, and trimmed off if they exist, to avoid their interference
on subsequent alignment and other steps. Additional data cleanup is carried
out after the alignment step.

8.3.2 Alignment and Transcript Counting


Alignment to the reference genome is a key preprocessing step prior to most
other preprocessing steps. Aligners used for bulk RNA-​seq, such as STAR
and the later aligner kallisto [28], can be directly applied to scRNA-​seq
data. As covered in Chapter 7, STAR is a widely used RNA-​seq aligner that
employs a “seed-​and-​extend” strategy for ab initio splice junction detection.
For aligning scRNA-​seq reads, the aforementioned Cell Ranger and zUMIs
use STAR as their aligner of choice. While STAR is currently one of the top
performing methods, especially in terms of accuracy and alignment rate, it
is relatively slow in speed and demanding on computational resources. To
achieve faster speed with less computer memory, kallisto is developed to
use a de Bruijn-​based process called pseudoalignment. Instead of mapping
sequencing reads, it is designed to map k-​mers extracted from reads to speed
up the process (Chapter 7, Section 7.3.2). Other bulk RNA-​seq alignment
methods covered in Chapter 7, including BWA, Bowtie2, and TopHat2, may
also be used for mapping scRNA-​seq reads.
After alignment, reads can be classified as exonic, intronic, or intergenic
for the purpose of counting transcripts. Different pipelines may use slightly
different definitions for this classification process. For example, Cell Ranger
considers a read to be exonic if at least 50% of the sequence intersects an
exon. Traditionally, only exonic reads are counted. When snRNA-​seq is used,
however, intronic reads derived from nascent transcripts, or transcripts that
were in the middle of being spliced, can also be informative and counted.
To perform transcript counting, for platforms that incorporate cell-​specific
barcodes, such as 10× Chromium, sequencing reads need to be assigned to
their cells of origin using the cell barcodes they carry. The 10× platform has
a “whitelist” of all GEM barcodes to help track cells. All reads associated
with a particular barcode are grouped together and considered to come from
one cell. As sequencing errors may exist in actual cell barcode reads, this
grouping process can be specified to tolerate such errors (e.g., one base devi-
ation from the whitelisted barcodes). For platforms that incorporate UMIs,
reads carrying the same UMI within the same cell are expected to be PCR
duplicates, and therefore need to be collapsed. Similarly, sequencing errors
in the UMIs may also be tolerated during collapsing of reads. Depending on
the method used, the collapsing of UMIs may be based on the UMI sequence
alone, or a combination of reads’ alignment location and the UMI sequence.
After the assignment of reads to cell barcodes and collapsing of reads using
Transcriptomics by Single-Cell RNA-Seq 157

Cell 1 Cell 2 Cell 3 . . . Cell N

Gene 1 1 220 23 ... 150

Gene 2 0 0 3 ... 1
Gene 3 5 2 13 ... 0

...
...

...
...
...
...

Gene M 0 18 12 ... 0
FIGURE 8.4
Gene-​cell count matrix.

the UMIs, the number of UMIs is counted for each gene and each cell barcode,
thereby generating a gene-​cell matrix (Figure 8.4). This matrix becomes the
basis of nearly all downstream analyses.
The above processes of genome alignment, reads classification, barcode
assignment, UMI collapsing, and transcript counting can be accomplished
together in a single workflow using various pipeline tools. For instance, for
10× data the “cellranger count” pipeline is typically used. However, it requires
significant computational resources and is not very fast. To increase pro-
cessing efficiency and speed up these preprocessing steps, newer tools such as
STARsolo [29], kallisto/​bustools [30], and Alevin [31] have been developed.
Besides 10× data, these later tools can also be used to process scRNA-​seq data
generated from other platforms. As suggested by its name, STARsolo is built
around the high-​performing STAR aligner for read mapping, as well as cell
barcode assigning, UMI collapsing, and gene-​cell matrix creation. Kallisto,
which aims to achieve a balance between computing efficiency and accuracy,
can generate pseudoaligned scRNA-​seq data in a new format called BUS,
which provides a binary representation of the data in the form of barcode,
UMI, and sets of equivalence classes. Once generated, BUS files can be
manipulated with bustools [30] to produce a data matrix consisting of gene
count and barcode. To improve the accuracy of gene abundance estimates,
Alevin includes gene-​ambiguous reads in its quantification, i.e., those that
multimap between genes and are usually discarded by other tools.

8.3.3 Data Cleanup Post Alignment


For a high-​throughput platform such as 10× Chromium, additional data
cleanup is needed post alignment to remove signal associated with doublets
(or multiplets), dead cells, or ambient RNA. On the 10× platform, not all
reads associated with GEM barcodes from the “whitelist” are derived from
bona fide single cells. As mentioned earlier, doublets (or multiplets) form
through partitioning of two (or more) cells into the same GEM. Dead cells
and ambient RNA molecules can also be partitioned into GEMs like authentic
158 Next-Generation Sequencing Data Analysis

cells. Detection and removal of reads associated with such unwanted GEMs
are based on three indicators: the number of uniquely mapped genes per
GEM barcode, the total count of UMIs per GEM barcode, and the fraction of
reads per barcode that map to the mitochondrial genome. If a barcode has
unusually high gene and UMI counts, it is possibly associated with a doublet
(or multiplet). Conversely, if a barcode is associated with few mapped genes,
a low UMI count, and/​or a high fraction of mitochondrial genes, it is an
indication of ambient RNA or a dead cell in the original sample. A dead cell
may have most of its cytoplasmic mRNA leaked out due to compromised
cell membrane, with only mitochondrial RNA preserved because of the
organelle’s double membrane system (Chapter 1, Section 1.4.9). To detect
such unwanted reads, these three quality indicators should be used in com-
bination rather than alone. It should also be noted that under some cellular
conditions these commonly used quality indicators may be violated. For
example, cells in high metabolic state may have unusually higher mitochon-
drial RNA fraction, or very large cells may appear to be doublets.
Doublets/​multiplets represent hybrid-​or super-​transcriptomes and com-
pound downstream data analyses if not removed. Besides the basic detec-
tion method using the total count of genes and UMIs as mentioned above, a
number of specially developed doublet/​multiplet detection tools are avail-
able, including DoubletFinder [32], Scrublet [33], DoubletDetection [34],
cxds and/​or bcds [35], and solo [36]. In general, these methods work by first
building artificial doublets from combination of randomly selected droplets
(or GEMs), then generating a “doublet score” for each droplet based on their
similarity to the artificial doublets, and finally calling doublets if the score
surpasses a threshold. The major difference between these methods is on
how the doublet score is generated. For example, DoubletFinder, one of the
top performing tools based on a benchmark study [37], uses the k-​nearest
neighbors (kNN) method to calculate the proportion of nearest artificial
doublet neighbors as the score for each droplet. It should be noted that none
of these tools works well for every case. In addition, to avoid removing large
sized cells that appear to be doublets, the rate of doublets/​multiplets identi-
fied by these tools for removal should not exceed that expected from Poisson
statistics for the experimental condition.
Besides using software tools alone, experimental techniques can also be
used to improve their detection, such as cell hashing [38], or mixing of cells
from different species (e.g., human and mouse cells). With cell hashing,
antibodies against ubiquitous cell surface proteins are tagged with oligo-
nucleotide barcodes to distinguish cells from different samples for robust
identification of cross-​ sample doublets/​ multiplets. Along the same line,
genotypic differences between cells, such as those collected from unrelated
individuals, can also be used to for detection of doublets/​multiplets. For
example, tools like demuxlet [39], scSplit [40], souporcell [41], and Vireo [42]
can separate mixed cells into individual samples based on each sample’s
Transcriptomics by Single-Cell RNA-Seq 159

unique SNP profile, and thereby identify doublets/​multiplets if mutually


exclusive SNP profiles are associated with the same droplet. Among these
tools, demuxlet requires knowledge of genotypic differences between indi-
vidual samples a priori, while the later tools (scSplit, souporcell, and Vireo)
infer genotypic differences from the observed scRNA-​seq reads.
To keep the rate of doublets/​multiplets low, the majority of droplets are
empty without any cells. However, ambient RNA molecules in the cell
sample buffer, if exist, can still be captured by empty droplets as well as those
containing cells. Reads generated from droplets associated with only ambient
RNA do not represent real cells and need to be removed. Since only a small
number of genes and UMIs are expected to be detected in these droplets, they
can be identified based on their low gene and UMI counts and the associated
reads filtered out. This filtering approach, however, may also remove small
cells that have low RNA content. To overcome this problem, tools such as
EmptyDrops [43], dropkick [44], and CB2 [45] can be used. These tools rely
on modeling of ambient RNA profile, against which the RNA profile of each
droplet is compared to determine whether it is associated with a genuine
cell. For example, EmptyDrops first constructs an ambient RNA profile using
droplets with low UMI counts that most likely represent empty droplets. For
droplets that are not associated with high UMI counts (those with high UMI
counts are automatically considered to represent cell-​containing droplets),
their RNA profiles are tested for deviations from the ambient RNA profile
using a Dirichlet-​multinomial distribution model [43]. Droplets with RNA
profiles that significantly deviate from the ambient RNA model are identified
as genuine cells, even if they have low UMI counts (Figure 8.5). This method
is incorporated into Cell Ranger from version 3.0, which leads to improve-
ment in identifying cells of low RNA content. Besides these tools, others such
as DecontX [46] and SoupX [47] have been developed to remove ambient
mRNA contamination in cell-​containing droplets to reveal their true gene/​
UMI counts.
The fraction of UMIs from the mitochondrial transcriptome is another
indicator of cell preparation and data quality. High-​quality cell preparations
should have minimal cell death and typically generate only low percentages
of reads of mitochondrial origin (<5% for cells of low energy demands, such
as adrenal, lung, and white blood cells) [20, 48]. Mitochondria have their
own small genome and transcriptome, but in a typical cell the amount of
mitochondrial RNA is relatively low compared to the large amount of RNA
transcribed from the nuclear genome. If the fraction of mitochondrial UMI
counts is significantly higher in a droplet, however, it is a strong indication
that it captured a dead cell. If a significant percentage of droplets is associated
with high mitochondrial reads, it is an indication of low cell sample quality. It
should also be noted that some cells, due to biological factors such as stress or
high metabolic activity (such as cardiomyocytes, hepatocytes, kidney cells),
may display higher mitochondrial gene/​UMI counts because of the presence
160 Next-Generation Sequencing Data Analysis

All barcodes Empty droplets


10000

10000
1000

1000
Total count

Total count
100

100
10

10
1

1
1e+00 1e+02 1e+04 1e+06 1e+00 1e+02 1e+04 1e+06
Rank Rank
Large cells Small cells
10000

10000
1000

1000
Total count

Total count
100

100
10

10
1

1e+00 1e+02 1e+04 1e+06 1e+00 1e+02 1e+04 1e+06


Rank Rank
FIGURE 8.5
Detection of empty droplets vs. those containing cells. Total UMI count is plotted against the
rank for each barcode in a scRNA-​seq dataset from Lun et al. (2019). Plots are shown for all
barcodes, and those associated with empty droplets, large and small cells, respectively. Using
this method (EmptyDrops) barcodes associated with small cells can be distinguished from
those with empty drops. (Adapted from: ATL Lun, S Riesenfeld, T Andrews, TP Dao, T Gomes,
et al., EmptyDrops: distinguishing cells from empty droplets in droplet-​based single-​cell RNA
sequencing data, Genome Biology 2019, 20(1):63. Used under the terms of the Creative Commons
Attribution 4.0 International License, https://​crea​tive​comm​ons.org/​licen​ses/​by/​4.0, ©2019
Lun et al.)

of larger numbers of mitochondria and/​or increased mitochondrial gene


expression [48].

8.3.4 Normalization
While the use of UMIs removes the effects of PCR duplicates, other factors
during single-​cell sample processing can still introduce undesirable variations
among cells or samples. Such factors include RNA capture rate, reverse tran-
scription efficiency, random sampling of molecules during sequencing, and
sequencing depth. If uncorrected, such variations may lead to inaccurate
results in downstream analytic steps. The goal of normalization is to correct
Transcriptomics by Single-Cell RNA-Seq 161

for such variations in order to make cells and samples directly comparable.
Normalization approaches developed for bulk RNA-​seq (Chapter 7, Section
7.3.3) may be used for scRNA-​seq data, especially those generated on low-​
throughput platforms with full-​length transcript coverage. In general, how-
ever, normalization of scRNA-​seq data faces unique challenges mostly due to
the issue of signal sparsity. To address the challenges, a number of methods
have been developed specifically for normalizing scRNA-​seq data. Examples
of these methods are SCnorm [49], Linnorm [50], BASiCS [51], Census [52],
ZINB-​WaVE [53], and sctransform [54]. Besides these dedicated normal-
ization methods, commonly used scRNA-​seq pipeline toolkits such as Cell
Ranger, Seurat [55], Scanpy [56], scran [57], and scVI [58] also contain their
own normalization methods.
These normalization methods can be generally classified into two general
categories: global scaling-​based and modeling-​based. The former approach,
represented by BASiCS and those employed by Cell Ranger, Seurat, Scanpy,
and scran, is based on the use of a global “size factor.” As an example, Cell
Ranger performs normalization in its aggr pipeline, through subsampling
of libraries of higher sequencing depth until all libraries have on average
the same number of confidently mapped reads per cell. Seurat, as another
example, includes a similarly simple normalization process performed as
follows: first divide the UMI count of each gene by the total number of UMIs
in each cell, then multiply the resultant ratios with a scaling factor (typic-
ally in the range of 104 to 106), and lastly perform a log transformation of the
scaled values (actually log(x+​1) to accommodate zero count genes) to gen-
erate normalized data. This global scaling approach assumes that RNA con-
tent is constant across all cells, and that one size factor fits all genes/​cells. This
assumption may not be true at times, especially with highly heterogeneous
population containing cells of different sizes and RNA content. To address this
concern, methods in the other category, as represented by SCnorm, Linnorm,
Census, ZINB-​WaVE, sctransform, and scVI, are based on modeling of cel-
lular molecule counts using probabilistic approaches. SCnorm, for example,
employs quantile regression modeling, which is used to group genes based
on the relationship between their UMI count and sequencing depth. Genes
in different groups are normalized using group-​specific size factors. With
sctransform, which is included in Seurat from ver3 as a normalization option,
a regularized negative binomial (NB) regression model is used. This method
first constructs a generalized linear model (GLM) for each gene to estimate
the relationship between its UMI count and sequencing depth. The estimated
parameters are then regularized based on gene expression level. To generate
normalized values for each gene, the regularized parameters are applied to
an NB regression model. Overall, these methods are based on models that
make different assumptions about the sparsity and underlying distribution
of gene expression values in cells. Comparative studies on these methods
have reported that the performances of different methods vary from dataset
162 Next-Generation Sequencing Data Analysis

to dataset, and therefore it is advisable to use more than one method on the
dataset at hand and then select the one that has the best performance [59, 60].
Variance stabilization is often an inherent goal of normalization. The log
transformation used in many methods toward the end achieves this goal
while also makes the normalized gene expression approximate a normal
distribution to facilitate downstream analyses. Without this stabilization,
the magnitude of a gene’s average gene expression correlates with the mag-
nitude of its variance, i.e., the so-​called mean-​variance relationship. Based
on this relationship, highly expressed genes also tend to have high levels
of variance, even if they do not contribute to cellular heterogeneity, such as
housekeeping genes. Conversely, genes that are expressed at low levels have
relatively low variance, even if they are biologically significant including
those coding for transcription factors. The goal of variance stabilization is to
remove the unwanted effects of this relationship, so that genes with greater-​
than-​expected variance between cells, regardless of the magnitude of their
expression, can be revealed.

8.3.5 Batch Effects Correction


If samples must be divided into batches, batch effects correction needs to be
performed. If left uncorrected, technical variations between batches caused
by factors such as different reagent lots, operators, protocols, sequencers, or
flow cells/​lanes will compound data analysis and affect results. Batch effects
can be detected through exploratory data analysis, such as visual inspection
of cells in a low-​dimensional space (Figure 8.6; more details on data visualiza-
tion can be found in Section 8.4.3). To correct for batch effects between highly
similar biological replicates in an experiment, where the technical variations
between batches are mostly due to the use of different flow cells and sequen-
cing depth, a simple approach is to regress them out. Implemented in pipe-
line tools such as Seurat and Scanpy, this approach uses linear regression to
regress out the unwanted effects of experimental batches and other technical
covariates, as well as biological covariates (such as cell cycle). Tools that are
specially designed for batch effects correction, such as ComBat, have also
been shown to work well [61]. ComBat, originally developed for correcting
batch effects in bulk gene expression analysis [62], uses a linear regression
model to model normalized gene expression with experimental batch as a
covariate. It first generates batch-​specific mean and variance for each gene,
and then an empirical Bayes-​based adjustment is applied to all genes to
remove batch effects, thereby generating a batch corrected expression matrix
as output.
For data integration that involves datasets generated from different
experiments or laboratories, where technical variations can be due to more
diverse factors including different cell preparatory procedures, sequencing
setups, or even overall approaches and platforms, the above methods for
Transcriptomics by Single-Cell RNA-Seq 163

No batch correction Batch correction

FIGURE 8.6
Correction of batch effects. Without batch correction (left) batch effects are evident with
cells being colored by their original batches. With batch correction (right) batch effects are
removed. (Adapted from: MD Luecken, FJ Theis, Current best practices in single-​cell RNA-​
seq analysis: a tutorial, Molecular Systems Biology 2019, 15(6):e8746. Used under the terms of
the Creative Commons Attribution 4.0 License, https://​crea​tive​comm​ons.org/​licen​ses/​by/​4.0,
©2019 Luecken et al.)

intra-​experiment batch effects correction may underperform. Here methods


that can be used include fastMNN [63], Seurat Integration [64], LIGER [65],
Scanorama [66], BBKNN [67], Harmony [68], and Conos [69]. The fastMNN
method is one implementation of the mutual nearest neighbors (MNN)
approach, which is based on generalized non-​linear modeling. MNN detects
pairs of cells in two datasets that are most similar to each other (mutual
neighbors) based on their gene expression profiles and therefore assumed to
be equivalent cell types. Assuming the batch effects are orthogonal to the bio-
logical subspace, the systematic differences between the cells in all the MNN
pairs are used to estimate the direction and magnitude of the batch effects,
which is then applied to transform the original data of all cells to remove
the batch effects. Seurat Integration, as implemented in Seurat 3, also uses
the MNN approach to detect equivalent cell types, but this is performed in a
subspace created by another algorithm called Canonical Correlation Analysis
(CCA) [55], which aims to find shared sources of variation between datasets.
The CCA process starts with identifying linear combinations of genes that
have maximum correlation between datasets. This is followed by aligning
the resultant vectors across the datasets using a nonlinear transformation
process called dynamic time warping, thereby creating a low-​dimensional
subspace containing all datasets prior to applying the MNN procedure.
Scanorama is based on a similar approach performing searches for MNNs
164 Next-Generation Sequencing Data Analysis

in a low-​dimensional subspace created by transformation of the original


gene expression using a method called singular value decomposition (SVD).
The Chemistry Batch Correction algorithm used by Cell Ranger since v3
for correcting batch effects between chemistries is also based on the MNN
approach.
Besides the MNN-​based methods above, BBKNN applies the kNN algo-
rithm to remove batch effects from diverse datasets and create batch balanced
data. The kNN procedure first constructs a cell neighborhood graph to iden-
tify the k nearest neighbors for each individual cell within each batch of data.
This is followed by merging of the nearest neighborhood sets across different
batches to connect equivalent cell types, while still keeping dissimilar cell
types separated. Through creating connections between equivalent cell types
from different datasets batch effects are quantified and corrected. LIGER,
or Linked Inference of Genomic Experimental Relationships, offers another
approach that has been shown to perform well on integrating different
scRNA-​seq datasets [70]. It employs a method called integrative non-​negative
matrix factorization (iNMF) to identify shared and dataset-​specific factors.
The low dimensional shared factor space is then used to identify similar cell
types across datasets, and joint cell clusters for batch correction. All of the
dataset integration methods introduced above aim mostly to correct batch
effects between technically more variable datasets collected from different
experiments or labs. If they are used for intra-​ experiment batch effects
correction, they might lead to over-​correction. To determine how a batch
effects correction method has performed, visualization of the data before and
after the correction in a low-​dimensional space should offer a quick qualita-
tive assessment (Figure 8.6). For a quantitative assessment, metrics such as
kBET [61] and average silhouette width (ASW) [71] can be used.

8.3.6 Signal Imputation
As mentioned at the beginning of this chapter, technical factors such as low
mRNA molecular capture rate and non-​exhaustive sequencing cause signal
dropout, i.e., the inability to detect a transcript that is present in a cell. This
leads to high signal stochasticity, signal sparsity, and zero inflation, all of
which can affect downstream data analyses. Signal imputation, also called
denoising or expression recovery, aims at inferring missing transcript values
to help alleviate this problem. Some of the commonly used scRNA-​seq signal
imputation methods include MAGIC [72], kNN-​smoothing [73], SAVER and
SAVER-​X [74, 75], ALRA [76], CIDR [77], DCA [78], scImpute [79], mcImpute
[80], and DrImpute [81]. Some of the pipeline tools introduced earlier have
built-​in imputation function (such as scVI), or use wrappers to run exter-
nally developed imputation tools (such as Seurat running imputation using
ALRA). These methods can be separated into three groups based on the
general approach they employ [82]: (1) modeling-​based: SAVER, CIDR, and
Transcriptomics by Single-Cell RNA-Seq 165

scImpute, for example, use probabilistic models to quantify signal uncer-


tainty from sparse count data to differentiate technical zeroes (for which
imputation will be provided) from biological zeroes (no imputation needed);
(2) data-​smoothing-​based: MAGIC, kNN-​smoothing, and DrImpute adjust
all expression values, not just zeros, based on gene counts in similar cells; and
(3) data-​reconstruction-​based: ALRA, DCA, mcImpute, and scVI use machine
learning or low-​rank matrix methods to create a latent space representation
of the original data to reconstruct the cell-​gene count matrix.
While signal imputation can lead to increased performance in downstream
analyses, it should also be noted that these tools have the inherent limitation
of being “circular,” i.e., inferring missing transcript values using information
from within the original data. As a result, they run the risk of generating
false-​positive results [83]. Benchmark studies also show that the performance
of these methods varies with the type of downstream analyses performed
[81, 84]. There is currently no consensus on whether signal imputation is
an essential preprocessing step, largely due to the difficulty in assessing
imputed data. The reader should be cautious on applying signal imputation
techniques on normalized or raw data. If performed it is prudent to visually
inspect and, better yet, experimentally validate results produced from the
imputed data. Like in the case of genotype imputation [85], use of a reference
database of single-cell transcriptomic profiles, such as the Human Cell Atlas,
may help improve imputation performance. In this direction there are some
emerging methods, such as SAVER-​X, that use information from reference
database or external dataset for transfer learning.

8.4 Feature Selection, Dimension Reduction, and Visualization


8.4.1 Feature Selection
Among the many genes contained in a dataset, many of them may not be very
informative because, for example, their expression remains constant across
cells (such as housekeeping genes). While it is not mandatory to preclude
these genes from further analysis, including them leads to high memory
usage and slow processing speed. To increase computational performance,
feature selection approaches can be employed to identify the most biologic-
ally informative features (i.e., genes, with their numbers usually in the range
of 500 to 2,000), and remove uninformative features for downstream ana-
lyses such as dimension reduction and visualization. Uninformative genes
can be selected based on their expression levels, with those showing zero
or low expression often being the target. This approach only needs to set
a threshold on the level of mean gene expression across all cells. The most
informative genes are often those that display high expression variability
166 Next-Generation Sequencing Data Analysis

between different cell groups or identities, and therefore most likely con-
tribute to cell-​to-​cell variation. This relies on the premise that genes showing
high variability across cells are a result of biological effects, not experimental
noise. Methods for selecting highly variable genes (HVGs) include the
squared coefficient of variation method proposed by Brennecke et al. (2013)
[86], the FindVariableGenes method used by Seurat, or those incorporated in
other pipeline tools scran and scLVM [87]. Because of the heteroscedasticity
of cellular gene expression, i.e., the aforementioned mean-​variance relation-
ship, the selection of HVGs should not be based on variance alone. Instead,
these methods fit the relationship between variance and the mean into their
respective models, based on which HVGs are selected using different stat-
istic tests. For example, scran performs LOESS fit on the variance-​mean
relationship, and then uses the fit as the model to infer biological variation
across cells for HVG selection. Evaluation of commonly used HVG selection
methods reported large differences among the methods, and that different
tools perform optimally for different datasets [88]. Besides using the vari-
ability of a gene’s expression across cells, alternative feature selection strat-
egies include using average gene expression level to select genes with the
highest average expression [89], deviance to identify genes that deviate from
the null model of constant expression [90], or dropout rate to retain genes
with higher number of dropouts than expected [91].

8.4.2 Dimension Reduction
Collected from a large number of cells (e.g., tens of thousands) for a large
number of genes (potentially all genes in the genome), scRNA-​seq data
has high dimensionality. This, in data science terms, causes the curse of
dimensionality, i.e., the amount of data needed for accurate generalization
grows exponentially with the increase in dimensions. Computationally,
high dimensionality leads to mathematical intractability for many modeling
and statistical calculations. Feature selection is one preliminary step toward
dimensionality reduction. However, even after this step the dimensionality
of the data is still very high. For example, there are still 2,000 gene dimensions
if the top 2,000 HVGs are retained. For many downstream analytical steps,
such as visualization and clustering to be detailed next, the number of
dimensions must first be significantly reduced to only a very small number
of dimensions (e.g., 10). To achieve this, specialized dimensionality reduc-
tion methods are required. The goal of these methods is not to select a small
number of original features, but to transform the data to create new features
so that the information contained in the original data can be preserved in the
low-​dimensional space.
Among the most commonly used dimensionality reduction methods are
those based on linear transformation. Principal components analysis (PCA),
independent component analysis (ICA) [92], non-negative matrix factorization
Transcriptomics by Single-Cell RNA-Seq 167

(NMF) [93], and factor analysis [94] are all examples that have been applied
to scRNA-​seq data. Among these methods, PCA is perhaps the best known.
It projects the cell-​gene count matrix onto a subspace that is defined by a few
principal components, which are linear combinations of the original genes.
The first principal component, or PC1, is one axis in the new subspace along
which the maximal amounts of variation in the original data are captured.
The second axis, corresponding to PC2, is orthogonal to the first axis catching
the second most variation in the data. Although straightforward, PCA does
not take signal dropout into consideration. ZIFA (zero-​inflated factor ana-
lysis), often considered to be a variation of PCA, is developed to address this
issue [94]. Although effective, such PCA-​based approaches have one down-
side, i.e., the principal components that catch the majority of variance in the
original data are sometimes difficult to interpret biologically. Other methods,
such as f-​scLVM (or factorial single-​cell latent variable model) and NMF,
address this difficulty through generation of reduced dimensions that are
more biologically relevant. For example, reduced dimensions from f-​scLVM
are based on explicit modeling of bio-​pathway annotations of gene sets [95].
The linear dimensionality reduction methods introduced above are based
on the assumption that the underlying data structure is linear in nature.
Methods that are based on non-​ linear transformation do not make this
assumption, instead these methods operate under the premise that in a high-​
dimensional space most relevant information concentrates in a small number
of low-​ dimensional manifolds. Currently available non-​ linear methods
include t-​ SNE (t-​distributed Stochastic Neighbor Embedding) [96], MDS
(Multi-​Dimensional Scaling) [97], Isomap (Isometric Feature Mapping) [98],
LLE (Locally Linear Embedding) [99], diffusion maps [100], spectral embed-
ding [101], and UMAP (Uniform Manifold Approximation and Projection)
[102, 103]. The t-​SNE algorithm, for example, works by modeling transcrip-
tionally similar cells around each cell based on probability distribution,
with Gaussian and t-​distributions being used in the original and dimension-​
reduced space, respectively. The process first computes cell-​cell similarity
in the original space using a Gaussian kernel and then maps the cells to a
dimension-​reduced space that best preserves that similarity. The strength of
t-​SNE is to reveal local data structure, but this is often achieved at the expense
of global data structure. An important parameter in the t-​SNE algorithm is
perplexity, which effectively controls the number of transcriptionally similar
cells that each cell has. Proper adjustment of this parameter can help regulate
the balance between local and global data structure [104]. UMAP is a more
recent algorithm that is designed to provide better preservation of global
data structure without losing performance on local data structure, better
scalability, and faster computation speed. While it has a similar procedure to
t-​SNE in that it first constructs a high-​dimensional representation of the ori-
ginal dataset and then projects it to a low-​dimensional space, UMAP differs
from t-​SNE in how cell-​cell similarity and the topology of cellular relations in
168 Next-Generation Sequencing Data Analysis

the original high-​dimensional space are computed and represented. UMAP


achieves these tasks through building the so-​called Čech complex, which is
essentially a mathematically efficient way of representing the topological
structure of cell-​cell similarities embedded in the original data structure.
UMAP has two key tunable parameters, i.e., the number of nearest neighbors
a cell can have in the high-​dimensional space, and the minimum distance
between cells in the output low-​dimensional space. By adjusting their values,
UMAP can reach a balance between local and global structure based on pro-
ject needs.
Other classes of dimensionality reduction methods include those based
on the use of autoencoders and those that use an ensemble approach. An
autoencoder, a type of unsupervised artificial neural network (ANN), aims
to compress a dataset to a lower dimensional space and then reconstruct it. It
achieves this by searching for representation of the dataset in the dimension-​
reduced space by focusing on key distinguishing features and removing
noise and redundant information. Methods using autoencoders include
VAE (Variational Autoencoder) [105], DCA (Deep Count Autoencoder) [78],
scvis [106], scScope [107], and scVI. SIMLR (single-​cell interpretation via
multikernel learning) is a method that uses the ensemble approach. It uses
t-​SNE for dimension reduction, but instead of using the Gaussian-​based cell
similarity measure as input to t-​SNE, it uses a cell similarity measure learned
from the multikernel learning framework to improve scalability and per-
formance of downstream steps [108].
Among the various dimensionality reduction methods introduced above,
benchmark comparisons [98, 109] show that no method excels for all datasets.
Non-​linear and autoencoder-​based methods can have better performance
on stability and accuracy especially for highly heterogeneous datasets, but
to achieve the performance users need to adjust parameters as parameter
settings can have significant impacts on results. Linear methods (such as
PCA) and UMAP tend to perform better in terms of computing speed, and
scalability especially when there are a large number of cells and genes in
the dataset. In practice, because of their efficiency, linear methods especially
PCA are often used to reduce dimensionality first, followed by visualization
of the resultant data using some of the non-​linear methods such as t-​SNE
and UMAP.

8.4.3 Visualization
One goal of dimensionality reduction is to enable graphical depiction of the
underlying data so that the researcher can visualize the major cell types (or
conditions) in the dataset, and thereby intuitively understand the inherent het-
erogeneity of the represented cellular population. In such a visual (Figure 8.7),
scatter plots are often used, in which each point represents a cell projected
into a two-​or three-​dimensional space, with each dimension corresponding
newgenrtpdf
Force-directed

Transcriptomics by Single-Cell RNA-Seq


PHATE PCA Diffusion maps t-SNE Isomap layout UMAP Monocle2
a Basophil C13 Erythrocyte C4
Dendritic cells C11 Erythrocyte C5
Early basophil C12 Erythrocyte C6
Paul (2015)
(n = 2,730)

Early erythrocyte C7 Lymphoid progenitors (NK) C1 9


Early monocyte C10 Megakaryocyte C8
Early neutrophil C9 Monocyte C14
Eosinophil C18 Monocyte C15
Erythrocyte C1 Neutrophil C16
Erythrocyte C2 Neutrophil C17
Erythrocyte C3

b 17 BC1A BC5D
18 BC1B BC6
Shekhar (2016 )

19 BC2 BC7
(n = 6,174)

21 BC3A BC8/9_ 1
23 BC3B BC8/9_ 2
24 BC4 Cone PR
25 BC5A Muller Glia
26 BC5B Rod BC
Amacrine_ 1 BC5C Rod PR
Amacrine_ 2

c
Astrocytes ependyma l
Zeisel (2015 )

Endothelial–mural
(n = 3,005)

Interneurons
Microglia
Oligodendrocytes
Pyramidal CA 1
Pyramidal SS

d 25
Embryoid body
(n = 16,825 )

20

15

Day
10

0
e 20.0
17.5
Zunder (2016 )

15.0
(n = 220,450)

12.5

Day
10.0
7.5
5.0
2.5
0

FIGURE 8.7
Visualization of single-cell RNA-​seq data in 2-​D space. Different visualization methods are applied to different datasets to show how these methods
differ from each other in generating visualizations for datasets of different characteristics. (Adapted by permission from Springer Nature Customer
Service Centre GmbH: Springer Nature, Nature Biotechnology, Visualizing structure and transitions in high-​dimensional biological data, Kevin R. Moon

169
et al., Copyright 2019.)
170 Next-Generation Sequencing Data Analysis

to the top reduced dimensions, or principal components if PCA was used


for dimension reduction. The actual rendering of the visualization from the
dimension reduced data is mostly achieved by non-​linear methods t-​SNE
and UMAP, which have better performance than other non-​linear methods.
Although PCA as the primary dimensionality reduction method may also be
used to visualize global data structure, and as a linear approach the distances
between cells or clusters in a PCA plot provide consistent depiction of their
relationships, its disadvantage as a visualization tool is its relative lack of
power to provide sufficient separation between distinct cell types.
As a non-​linear approach, t-​SNE typically offers a better presentation of the
same dataset, by placing similar cells close to each other and dissimilar cells
distant. As indicated earlier, using this method the similarities between cells
are measured locally based on their transcriptomic profiles. Because of its
emphasis on local similarity, t-​SNE tends to have limited preservation of the
dataset’s global structure. Although the perplexity parameter can be increased
to reveal more global structure, this usually leads to dramatically increased
computational time. It should be noted that even at low perplexity values
t-​SNE is not a fast method. Because t-​SNE emphasizes local structure and
may overestimate the differences between cells or cell clusters, the researcher
should be cautioned not to over-​ interpret the distance between cells or
clusters, the size of clusters, as well as the relative position of cells or clusters
in a t-​SNE visualization. As t-​SNE is a stochastic algorithm, the final output
may not be fully reproducible between runs. While revised implementations
of the t-​SNE approach aim to overcome such shortcomings [104], new visu-
alization methods, with UMAP being a good example and gaining popu-
larity, have been established as robust alternatives [102]. Compared to t-​SNE,
UMAP is faster, more scalable to very large datasets in terms of both data
size and dimensionality, and able to preserve more global structure while
retaining local data structure. While it is also stochastic, UMAP’s run-​to-​run
performance is generally more reproducible than that of t-​SNE. It should be
noted that as in the case of t-​SNE the distance between cells or clusters in a
UMAP visualization may not have biological bearing.
Besides t-​SNE and UMAP, other visualization tools are also available.
Example of these tools are PAGA (or partition-​ based graph abstraction)
[110], PHATE (or potential of heat diffusion for affinity-​based transition
embedding) [111], SPADE (or spanning-​tree progression analysis of density-​
normalized events [112], and SPRING [113]. Compared to t-​SNE and UMAP,
which are better for visualizing distinct groups of cells in a mixed population,
these other tools are more suited to display cells along a continuous trajec-
tory, such as those that are undergoing differentiation. Among these tools
PAGA provides a manifold learning algorithm that can faithfully preserve
both continuous and distinct cell identifies/​groups at multiple resolutions.
It is robust in terms of computational efficiency, scalability, and interpret-
ability of results for continuous cell trajectories (Section 8.7 covers “Trajectory
Inference”).
Transcriptomics by Single-Cell RNA-Seq 171

8.5 Cell Clustering, Cell Identity Annotation,


and Compositional Analysis
8.5.1 Cell Clustering
Based on their gene expression profiles, cells in a dataset can be grouped into
clusters. By placing transcriptionally similar cells into clusters, the process can
uncover different cell groups or identities in a sample, achieving a common
goal of scRNA-​seq analysis. This process often starts with dimension-​reduced
data, usually from PCA. To capture most of the gene expression variation
across cells in the dataset, enough principal components should be used. In
the dimension-​reduced space, different metrics can be used to quantify the
similarity (or distance) between cells based on their transcriptomic profiles.
Such similarity metrics can be mostly classified as distance-​or correlation-​
based. Two examples of distance-​based metrics are Euclidean distance, which
represents the shortest or straight-​line distance between two points (cells),
and Manhattan distance (also called city-​block distance), measuring the dis-
tance between two points in a grid like path. Correlation-​based metrics, such
as Pearson and Spearman’s correlation coefficients, measure the similarity
between the general shapes of gene expression in two cells. For comparison,
distance-​based metrics capture gene expression levels and measure how
far two cells are from each other in the PCA space, while correlation-​based
metrics track relative expression trends between two cells and are insensi-
tive to the scale of gene expression levels. A comparison of their clustering
performance showed that correlation-​based metrics are more robust than
distance-​based metrics [114]. Other similarity metrics have also been used,
e.g., cosine similarity, a measure of angle instead of magnitude between two
vectors, that is used for spherical K-​means clustering [115].
There are different algorithms to cluster cells, and among the widely used
are hierarchical, K-​means, and graph-​based clustering. These algorithms
identify major cell identities (encompassing different cell types, or different
states in the same cell type) present in a population as clusters, and assign
each cell to one of these clusters without relying on any a priori informa-
tion. Hierarchical clustering constructs a dendrogram in which different cell
clusters merge or split at different branches, creating a hierarchy. One pro-
cess to create such a hierarchy is agglomerative, or “bottom-​up,” in which
at the start each cell is assigned to its own cluster, and at each subsequent
iteration the most similar nodes are merged. This process recurs hierarch-
ically, until all cells are merged. The opposite process takes a divisive or
“top-​down” approach, which starts with all cells being in a single cluster,
and then splits it into two branches at each subsequent step, until each cell is
assigned to its own cluster. From the dendrogram, clusters can be obtained
through “cutting the tree” to generate the desired number of clusters, or at
desired height.
172 Next-Generation Sequencing Data Analysis

The K-​means clustering algorithm first requires the user to specify k, the
number of clusters. The process starts with all cells randomly assigned to
one of k clusters. Then the centroid of each cluster is determined, and each
cell re-​clustered based on their distance to each of the k centroids. This pro-
cess is reiterated until each cell’s cluster assignment no longer changes. The
third commonly used approach, graph-​based clustering, is based on graph
construction that connects cells to their nearest neighbors. In a kNN graph
(different from K-​means), for example, two nodes (cells) are connected by
an edge, if the distance from cell A to B is within the k-​th lowest distances
from A to other cells. The edge may have a weight assigned based on the
similarity between the cells. In a Shared Nearest Neighbor (SNN) graph,
an edge is weighted based on their proximity to each other, or similarity in
terms of the number of mutual neighbors the two cells share. After such a
graph is constructed, dense regions that contain a large number of highly
connected nodes can be detected as the so-​called communities, representing
distinct cell identities. Within each community (or cluster), cells are more
highly connected with each other indicative of high similarity, than those in
other communities. To partition cells into distinct communities, community
detection techniques, such as the Louvain and Leiden methods [116, 117], can
be used.
To carry out these clustering approaches, either general-​purpose clustering
methods or tools specifically designed for scRNA-​seq data can be used. As
examples of general-​purpose methods, the hclust() and kmeans() functions
in R can be directly used on PCA dimension reduced data to perform hier-
archical and K-​means clustering, respectively. Examples of tools specially
developed for clustering single cells include SC3 [118], pcaReduce [119],
CIDR, RaceID2 [120], SIMLR [108], and SNN-​Cliq [121] (Table 8.1). Many
pipeline tools, such as Seurat, Cell Ranger, Pagoda2 [122], and Scanpy, scran,
ascend [97], and SINCERA [123], also provide built-​in clustering functional-
ities. Seurat, for example, provides graph-​based clustering. This process first
constructs a kNN graph using Euclidean distance in the PCA space, with the
edges weighted by Jaccard similarity that measures the number of neighbors
they share. To find clusters in the graph, the Louvain community detection
method is then applied. Seurat clustering uses a user adjustable parameter
called resolution to control the number of clusters generated, with a higher
resolution (e.g., >1.0) leading to a larger number of clusters. Cell Ranger
provides a similar nearest neighbor graph-​based clustering approach, with an
additional step to merge clusters that show no differential gene expression. In
addition, it also offers K-​means clustering as another option. Benchmarking
studies on most of these clustering methods show that there is wide variation
in actual performance and poor concordance among them [89, 124]. Some
methods, such as Seurat, SC3, and Cell Ranger, showed better overall per-
formance than others. In terms of running times, Seurat also showed consist-
ently faster speed than most other methods.
Transcriptomics by Single-Cell RNA-Seq 173

TABLE 8.1
Single-​Cell Clustering Methods
Name Description Reference

SC3 (Single-​cell PCA +​K-​means +​hierarchical clustering. Uses [118]


consensus clustering) multiple parallel clustering parameter tests
to reach a consensus matrix by hierarchical
clustering
pcaReduce PCA +​K-​means +​hierarchical clustering. An [119]
agglomerative clustering process that integrates
PCA and hierarchical clustering
SIMLR (Single-​cell Dimensionality reduction +​K-​means. Learns [108]
interpretation via proper weights for multiple kernels, and
multi-​kernel learning) constructs a symmetric similarity matrix for
dimension reduction and clustering
SNN-​Cliq Graph-​based clustering. Constructs an SNN [121]
graph and then clusters cells using quasi-​clique
finding techniques
RaceID2 Pearson’s correlation distance matrix for all pairs [120]
of cells +​K-​means clustering
CIDR (Clustering PCA +​hierarchical clustering. Uses implicit [77]
through Imputation imputation to help alleviate the effects of
and Dimensionality dropouts, then performs hierarchical clustering
Reduction) on principal coordinates
Seurat PCA +​kNN graph-​based clustering Seurat [55],
Pagoda2 Pagoda2 [122],
Scanpy Scanpy [56],
Cell Ranger PCA +​graph-​based clustering, or K-​means [10]
clustering
scran PCA +​hierarchical clustering, or SNN graph-​ [57]
based clustering
ascend PCA +​hierarchical clustering [97]
SINCERA Z-​score/​trimmed mean normalization +​ [123]
hierarchical (default)/​consensus/​tight
clustering

8.5.2 Cell Identity Annotation


After placing cells into different clusters, the next question is usually what cell
type, or what cellular state, is represented by each cluster. To help answer this
question, comprehensive pipeline toolkits such as Seurat, Cell Ranger, Scanpy,
and scran have built-​in functions to identify marker genes that are uniquely
highly expressed in a cluster compared to other clusters. For example, in
Seurat the FindMarkers() function identifies candidate marker genes for each
cluster by differential gene expression analysis, which is conducted using the
default non-​parametric Wilcoxon rank sum test among other available tests.
174 Next-Generation Sequencing Data Analysis

Cell Ranger finds differential genes for each cluster using edgeR (covered in
Chapter 7), or sSeq, which is a modified NB exact test [125]. From the iden-
tified differentially expressed, candidate marker genes, manual annotation
can be used to identify cell type or state in each cluster, based on previously
characterized, canonical cell identity specific marker genes. For example,
GFAP (glial fibrillary acidic protein) gene expression is a marker of astroglial
cells in the brain, and CD79a and CD79b are markers for B cells. The identifi-
cation of cell type or state based on such classic gene markers is an extension
of the traditional method often used in the lab for cell identity recognition.
This process, however, is typically labor-​ intensive and time-​ consuming,
as it often needs extensive review of currently available literature, or deep
domain knowledge about the cell system under study, which requires close
interactions between bench scientists and informaticians. In addition, some
cell types may not have well characterized gene markers, or the expression of
marker genes in host cells may be undetectable due to signal dropout.
To address some of the issues and speed up the cell identity recognition
process, a relatively easy and semi-​automatic approach is through Gene
Ontology (GO) and/​or bio-​pathway enrichment analysis of the candidate
marker genes (detailed in Chapter 7, Section 7.3.8), since the GO terms or
pathways significantly enriched in the genes can produce insights into cell
identity. For example, if “hepatocyte homeostasis” is identified as a signifi-
cantly enriched GO term, it is indicative that at least some of cells in the
cluster are hepatocytes. Further, the cell identity detection process on the
basis of canonical marker genes can be automated, with the use of specialized
tools such as Garnett [126], Digital Cell Sorter [127], SCINA [128], CellAssign
[129], and scANVI [130]. The list of marker genes required by these automated
tools to classify different cell identities can be provided by databases such as
PanglaoDB [131], CellMarker [132], the Mouse Brain Atlas [133], the BRAIN
Initiative Cell Census Network (or BICCN) [134], and DropViz.org [135].
Garnett, as an example, first uses as input a list of gene markers to train a
regression-​based classifier, which is then applied to classify cells in a new
dataset. As another example, scANVI, a semi-​supervised variant of scVI, also
classifies cells based on their expression of canonical marker genes. This tool
goes even further to use these cells as “seeds” to classify other cells in the
same dataset with unobserved expression of the marker genes, on the basis
of how close these other cells are to the “seeds.”
Instead of expression of marker genes, overall gene expression pattern may
also be used for automated inference of cellular identities. This approach uses
information embedded in the overall gene expression of annotated cells in a
reference dataset, to predict cell identities in a query dataset. Without reliance
on prior knowledge in the form of a pre-​defined list of marker genes, this
approach directly harnesses the power of rapidly accumulating single-​cell
data that can be used as reference, such as those from the Human Cell Atlas
[136] and the Tabula Muris Atlas for mouse [137]. Table 8.2 lists some of the
Transcriptomics by Single-Cell RNA-Seq 175

TABLE 8.2
Cell Identity Annotation Tools
Name Description Reference

Classifiers that do not require marker genes


scmap Mapping cells in a scRNA-​seq dataset onto the cell-​types or [138]
individual cells annotated in a reference dataset
SingleCellNet Uses a random forest classifier trained on annotated scRNA-​ [139]
seq data to classify cells in a query dataset
scPred Employs a combination of feature selection from a reduced-​ [136]
dimension space, and a support vector machine model to
classify single cells
ACTINN Uses a 4-​layer neural network for automated cell type [140]
identification
SingleR Assigns cell identify based on comparison of their [148]
transcriptomes to reference bulk transcriptomic datasets of
pure cell types
CHETAH Characterizes cell types by using correlation and confidence [149]
score in comparison to reference dataset using a hierarchical
classification tree
Cell BLAST Cell querying and annotation based on a neural network-​based [150]
generative model, and a posterior-​based cell-​cell similarity
metric
ItClust Iterative transfer learning with neural network to improve cell [151]
type classification
CaSTLe Classifies single cells based on feature selection and XGBoost [152]
classification
OnClass Uses Cell Ontology to embed different cell types into a low-​ [144]
dimensional space, and then maps cells to this partitioned
space based on gene expression for cell type annotation
CellO Performs hierarchical cell classification against the directed [145]
acyclic graph structure of the CO system
Classifiers that require marker genes
Garnett First trains a regression-​based classifier using the gene marker [126]
definition file, and then applies to classify cells from similar
tissues or sample types
SCINA Leverages established gene signatures to perform cell type [128]
classification
CellAssign Automates assignment of cells to known cell types using a [129]
probabilistic model with known marker genes as input
Digital Cell Assigns cell types automatically using a voting process based [127]
Sorter (DCS) on known molecular markers
scANVI A semi-​supervised classifier that uses Gaussian mixture model [130]
to provide posterior probability of assigning a cell to different
classes
176 Next-Generation Sequencing Data Analysis

methods that use this strategy. Among them, scmap [138] projects cells in
a query dataset onto a reference dataset (or combined references), to iden-
tify matching individual cells (the scmap-​cell mode) or specific cell types
(the scmap-​cluster mode). In this method, the similarity between individual
cells in the query sample and cells or cell types in the reference is measured
using three distance metrics (Pearson, Spearman, and cosine). Some of the
other methods are machine learning based, i.e., they first construct classi-
fier models from a reference dataset and then use the classifiers to annotate
individual cells or clusters from a query dataset. SingleCellNet (SCN), for
example, uses a random forest (RF) classifier trained on annotated reference
scRNA-​seq data to transfer to query cell classification [139]. ACTINN (or
Automated Cell Type Identification using Neural Networks) [140] is based
on neural network for such transfer learning. Another generalizable method,
scPred uses support vector machines (SVMs), combined with singular value
decomposition for unbiased feature selection, to perform probability-​based
cell type prediction [141]. Besides these machine learning-​based tools spe-
cially designed for single-cell data, general-​ purpose classifiers including
SVM and RF may also be used directly [142]. Among other cell classification
methods that use overall gene expression pattern instead of marker genes
are those developed for integrated analysis of multiple scRNA-​seq datasets,
such as Seurat Integration, Scanorama, and Conos as introduced in Section
8.3.5 (therefore not listed on Table 8.2). These methods achieve automated
annotation of cells from a query dataset through detection of equivalent cells
in a reference dataset. As an example, Seurat performs annotation of query
cells by mapping the query dataset onto a reference, which is accomplished
through projecting the query cells onto the reference UMAP structure.
Because the diverse array of methods introduced above may not always
use the same term to label the same cell type, for consistency cell annota-
tion can benefit from the use of standardized cell type terms from the Cell
Ontology (CO). CO is a community effort to organize cell types anatomic-
ally and hierarchically through a structured and controlled vocabulary [143].
To further leverage the inherent hierarchical relationships built into the CO
terms, specialized CO-​based cell annotation tools, such as OnClass [144] and
CellO [145], have also been developed. Cello, for example, performs hier-
archical classification to annotate cells based on the graph structure of the CO
system. Compared to methods that do not use CO terms, such tools provide
more consistent and standardized annotation of individual cells or clusters,
through the use of CO terms.
To evaluate the performance of these methods, comprehensive bench-
mark studies have been performed [146, 147]. Using 27 scRNA-​seq datasets
of varying cell numbers, platforms, species, and cellular heterogeneity, one
of the studies [146] compared 22 automated cell identification methods and
showed that the general-​purpose SVM classifier had the best overall per-
formance. Other top performers included SingleCellNet, scmap-​cell, scPred,
Transcriptomics by Single-Cell RNA-Seq 177

and ACTINN. After evaluating 10 methods on 6 datasets, the other study


[147] found Seurat to be the top performer (this method was not included in
the other study by Abdelaal et al.). SingleR and SingleCellNet also performed
reasonably well. Both studies found that methods that require marker genes
did not outperform those that use the overall gene expression pattern.
It should be noted that many of the methods evaluated by both studies
leave unknown cell types (i.e., those that display low or no expression of
all marker genes, or do not exist in the reference dataset) as unclassified,
which is desired. Such cells need to be further characterized using additional
canonical marker genes or biochemical techniques. In addition, many of the
evaluated methods, besides labeling distinct cell types/​identities, can also
be used to identify transitional or intermediate cell types as that they may
represent part of a developmental trajectory.

8.5.3 Compositional Analysis
The composition of different cell identities in a population varies with internal
and external conditions. For example, upon bacterial pathogen infection, in
the small intestinal epithelium there is a change in the proportions of different
cell types as part of antimicrobial response (Figure 8.8) [153]. Compositional
analysis involves examination of the proportions of different cell identities in
different samples. For this analysis, different statistical approaches have been
used to assess the significance associated with changes of cellular composition.
For example, to detect the change of cell composition in the intestinal epithe-
lium upon infection, Haber et al. (2017) applied a Poisson process to model

Control H. polygyrus (day 3)


H. polygyrus (day 10)
*
0.6 **
**
Fraction of tuft cells

0.4

0.2

0.0
Tuft Tuft-1 Tuft-2
progenitor
FIGURE 8.8
Changes in cellular composition in intestinal epithelium caused by pathogen infection. Shown
here are changes in the fraction of three different types of tuft cells (having chemosensory
function in the gut lining) after infection with the parasitic helminth Heligmosomoides polygyrus.
Significant changes in frequency are marked (* FDR < 0.25, ** FDR < 0.05; Wald test). (Adapted
by permission from Springer Nature Customer Service Centre GmbH: Springer Nature, Nature,
A single-​cell survey of the small intestinal epithelium, Adam L. Haber et al., Copyright 2017.)
178 Next-Generation Sequencing Data Analysis

the detected proportions of different cell types as a random count variable,


which used the grouping of each sample (treatment or control) as a covariate
and the total number of detected cells in each sample as an offset variable.
As another example, Hashimoto et al. (2019) used a Wilcoxon rank-​sum test
to compare the fractions of different classes of circulating lymphocytes in
supercentenarians vs. younger controls to uncover the mechanism of their
exceptional longevity through sustaining immune system function [154].
Single-cell differential composition, or scDC, is a specially developed tool to
assist such composition analysis. It employs a bootstrap resampling process
to determine the standard errors during estimating cell-​type proportions, and
generalized linear model (GLM) and mixed model (GLMM) analysis for subse-
quent significance testing to achieve statistical comparison [155].
One characteristic of compositional data is that they are subject to the
constant-​sum (i.e., unity) constraint [156]. This can lead to spuriously signifi-
cant correlations among different cellular identities and results in negative
bias. For instance, if a new cell type emerges due to activation, because of
the constraint the relative proportions of other existing cell types will cor-
respondingly decrease although their absolute numbers may not change.
Standard univariate analysis methods that do not take this characteristic into
consideration would lead to deflated estimation of existing cells. It is impera-
tive, therefore, to develop novel methods that account for such inherent bias
of single-cell compositional data. As an example, scCODA uses a Bayesian
approach that takes into consideration the negative correlative bias as well
as the uncertainty associated with determining cellular composition. It
achieves this through modeling cell type proportions using a hierarchical
Dirichlet-​Multinomial distribution, and joint modeling of all detected cell
type proportions instead of individual ones [157].

8.6 Differential Expression Analysis


The FindMarkers or other similar functions in Seurat, Cell Ranger, and
other tools for marker gene identification, as introduced in Section 8.5.2, are
in fact differential expression (DE) analysis methods that focus on finding
genes that are highly expressed in a particular cluster of cells vs. all others.
In this section, we focus on more general DE analysis to compare cells in
two (or more) populations, samples, time points, or perturbation conditions.
Methods developed for DE analysis of bulk RNA-​seq data, such as DESeq2,
edgeR, and limma, are also applicable to scRNA-​seq data. As detailed in
Chapter 7 (Section 7.3.5), DESeq2 and edgeR use the NB model under a GLM
framework, and Limma employs the normal linear model, to capture the
distribution of read counts. A core component of these methods is to assess
gene expression variance from the rather limited number of samples usually
used in bulk RNA-​seq, but in applying to scRNA-​seq data this is no longer a
Transcriptomics by Single-Cell RNA-Seq 179

challenge because of the much larger number of single cells involved in the
comparison. The substantial zero inflation, signal overdispersion, transcrip-
tional bursting, and multimodality associated with scRNA-​seq data, how-
ever, pose different challenges.
Methods developed for scRNA-​seq DE analysis (see Table 8.3) use different
approaches to address the specific challenges posed by single-cell data.
Examples of these methods are SCDE [158], MAST [159], D3E [160], scDD
[161], BPSC [162], NBID [163], DEsingle [164], DECENT [165], and SwarnSeq

TABLE 8.3
Single-​Cell Differential Expression Analysis Tools
Name Description Reference

Tools specifically developed for scRNA-​seq data


SCDE Uses a mixture of negative-​binomial distribution to model RNA [158]
signal amplification, and a Poisson distribution for signal dropout
MAST Models scRNA-​seq data using a mixture of Gaussian distribution [159]
to model gene expression level, and logistic regression to model
drop-​out events
BPSC Performs DE analysis based on a beta-​Poisson mixture model [162]
integrated into the GLM framework
DEsingle Adopts ZINB model to describe read counts and excessive zeros, [164]
and define and detect three types of DE genes
D3E Uses non-​parametric comparison of distributions for DE gene [160]
identification, and fits the transcriptional bursting model to
explore gene expression change mechanisms
scDD Uses a Bayesian framework to identify DE genes that are classified [161]
into different multimodal distributions
SwarnSeq Integrates ZINB and a binomial model to model UMI counts to [166]
account for zero inflation and RNA capture rates, to identify and
classify DE and differential zero-​inflated genes
Monocle 3 Conducts regression analysis to find genes that change expression [103]
under different experimental conditions, and/​or graph-​
autocorrelation analysis to identify genes that change with a
trajectory or differ between clusters
tradeSeq Performs trajectory based DE analysis using an NB generalized [167]
additive model
Tools originally developed for bulk RNA-​seq data
DESeq2 Models gene expression mean and variance using an NB [173]
distribution, and calculates each gene’s p value using the observed
sum of read counts of the two conditions
edgeR Uses an NB model to model gene expression, and performs DE [174]
analysis using the GLM likelihood ratio test
limma Fits a linear model for each gene, and use empirical Bayes [175]
procedures for borrowing information across genes for DE analysis
ROTS A general test originally developed for microarray data that uses an [176]
adaptive reproducibility-​optimized t-​like test statistic for DE analysis
180 Next-Generation Sequencing Data Analysis

[166]. To deal with zero inflation, SCDE fits a mixture of two error models,
with one using Poisson distribution to model the signal dropout process,
with the other using the NB distribution to model the signal amplification
process for detection of transcripts in correlation to their abundance in cells
[158]. MAST uses a two-​part generalized linear hurdle model, with one mod-
eling the discrete expression rate of each gene across cells (i.e., how many
cells express the gene) using logistic regression, and the other modeling the
continuous positive expression level of each gene by Gaussian distribution
[159]. To fit zero-​inflated and overdispersed scRNA-​seq data, SwarnSeq uses
the zero-​inflated negative binomial (ZINB) model to model the observed
UMI counts of transcripts. In addition, through using a binomial model to
adjust for cellular RNA capture rates, this method allows detection of DE
genes, as well as differential zero-​inflated genes, i.e., those that show signifi-
cant difference in the number of cells that have zero expression between
two groups [166]. To address the multimodality nature of scRNA-​seq data,
scDD employs Bayesian modeling to identify genes that display differential
distributions across conditions, and the genes are then further classified into
different multimodal expression patterns. SwarnSeq also classifies influential
genes into various gene types based on their differential expression and zero
inflation patterns. To address the issue of transcriptional bursting, D3E has
two modules, with one for DE gene identification, and the other for fitting a
model for transcriptional bursting to help discover the mechanisms under-
lying the observed expression changes.
The same marker gene finding functions contained in most compre-
hensive pipeline toolkits as introduced earlier can also be generalized
for DE analysis. For example, Seurat provides DE analysis from the same
FindMarkers() function through specifying two groups of cells for com-
parison. Currently available differential test methods include Wilcoxon
rank sum test, likelihood-​ ratio test, Student’s t-​ test, negative binomial
GLM, Poisson GLM, logistic regression, as well as the aforementioned
MAST and DESeq2. SINCERA, as another example, offers DE analysis using
one-​tailed Welch’s t-​test if gene expression can be assumed to come from
two independent normal distributions, or one-​tailed Wilcoxon rank sum
test in case of small sample sizes. To identify genes that are differentially
expressed along a developmental lineage or trajectory (Trajectory Inference
to be introduced next), methods such as Monocle 3 [103] and tradeSeq
[167] can be used. Monocle 3, for example, employs two approaches: graph
auto-​correlation and regression analysis. The former is suitable to identify
genes that change along a trajectory, or differ between clusters, while the
latter is to find genes that change expression under different experimental
conditions.
To evaluate the plethora of methods that are currently available for scRNA-​
seq DE analysis, several benchmarking studies were performed [168–​171].
Based on these studies, methods that were originally developed for bulk
RNA-​seq data have been shown to perform as well as methods developed
Transcriptomics by Single-Cell RNA-Seq 181

specifically for single-​cell data, especially after applying some strategies such
as prefiltering to remove lowly expressed genes [169] or weighting to deal
with zero inflation [172]. As these bulk or scRNA-​seq tools have different
ways of dealing with signal sparsity, multimodality, and heterogeneity, there
is a general lack of agreement in the DE genes they identify. In addition, these
benchmarking studies also show a general tradeoff between precision and
sensitivity, i.e., methods of high precision have low sensitivity, which leads
to identification of less true positive genes but also introduces fewer false
positives.
It should be noted that DE analysis is an integral step of an scRNA-​seq
analytical pipeline, and upstream data processing can have an effect on the
overall performance of this step. Of the various upstream steps, normaliza-
tion has been shown to have a significant impact on DE results by a system-
atic evaluative study conducted by Vieth et al. (2019). Based on this study, a
good normalization before DE analysis, such as that provided by scran, can
alleviate the need for complex DE methods [177]. Another note is that, just
like in bulk RNA-​seq analysis as detailed in Chapter 7 (Section 7.3.8), the
identified DE genes can be subjected to further functional analysis, such as
gene set enrichment analysis, to reveal what biological processes or pathways
are enriched in them.

8.7 Trajectory Inference
Many biological processes, such as development, immune response, or tumori-
genesis, are underlined by continuous dynamic cell changes across time. The
path of changes that a cell undergoes in such a process is often called a tra-
jectory. While it is not yet possible to monitor the continuous transcriptomic
change of an individual cell over time, trajectory can be inferred from a popu-
lation of cells that represent a continuum of transitional cellular states while
cells undergo changes in an unsynchronized manner. Because trajectory
inference (TI) is based off of a snapshot of gene expression of a population
of cells at a certain point of time, it is also called pseudotemporal analysis.
Methodologically, it is built on the premise that cells in the continuum share
many common genes and their gene expression displays gradual change. In
essence, to infer cellular trajectory is to find a path in the cellular gene expres-
sion space that connects cells of various transitional states by maximizing
similarity between neighboring cells. The inferred cellular trajectory can then
be validated with additional experimental evidence.
Trajectory inference is carried out on dimensionality-​reduced data, often
after the clustering step. General methods, such as minimum spanning tree
(MST) that aims to connect all points (clustered cells) in a graph to a path
that minimizes distance between points, can be directly used for TI [178].
182 Next-Generation Sequencing Data Analysis

Some of the methods specifically developed for TI are in fact based on MST.
For example, the first version of Monocle, a pioneer method for inferring tra-
jectory from single-cell sequencing data, first creates MST on cells projected
in a dimensionality-​reduced space, and then places cells along the longest
path through the MST [92]. Slingshot [179], TSCAN [180], and Waterfall [181]
build MST on cell cluster centroids, instead of cells, and then order cells onto
the path through orthogonal projection. Besides these MST-​based methods,
some other commonly used methods are based on graph theory. For example,
Diffusion Pseudotime (DPT) builds weighted kNN graph on cells, and then
orders cells using random-​walk-​based distance [182]. Also based on the use
of weighted kNN graph, PAGA performs graph partitioning and abstraction
using the Louvain method to identify different cellular states or identities,
and uses an extension of DPT for pseudotime calculation [110] (Figure 8.9
shows an example). Monocle 3 is built on PAGA and adds one step further
to construct more fine-​grained trajectory through learning a principal graph
from the PAGA graph [103].
The methods mentioned above and listed on Table 8.4 are among an increas-
ingly long list of TI methods available. Besides the different approaches these
methods use to infer trajectories, they also differ in what trajectory topology
they can infer, whether they require prior information, how scalable they
are with increasing cell numbers, etc. Cellular trajectory topologies can be

muscle

secretory muscle pharynx cell type


progenitors
epidermis DVb

epidermal
neoblast neural
progenitors progenitors
gut neuronal
epidermis progentors
parenchymal
progenitors

parenchymal gut

FIGURE 8.9
Cell trajectories inferred by PAGA to reconstruct a developmental lineage tree encompassing
all cell types in the planarian body based on single-cell transcriptomic data. (Adapted from FA
Wolf, FK Hamey, M Plass, J Solana, JS Dahlin, B Göttgens, N Rajewsky et al., PAGA: graph
abstraction reconciles clustering with trajectory inference through a topology preserving map of
single cells, Genome Biology 2019, 20(1):59. With permission.)
Transcriptomics by Single-Cell RNA-Seq 183

TABLE 8.4
Trajectory Inference Methods
Name Description Reference

PAGA Constructs a weighted kNN graph, followed by graph [110]


partitioning and abstraction using the Louvain method.
Applies an extended version of DPT for pseudotime
calculation
Slingshot Uses cell cluster-​based MST to learn global lineage structure, [179]
and then orders cells along lineage curves through orthogonal
projection
Monocle Initial version (Monocle 1) builds an MST on cells in an ICA [92, 103]
dimension reduced space and then orders cells along the
longest path. Monocle 3 learns principal graph from PAGA
graph to build more fine-​grained trajectory
TSCAN Constructs MST to connect cell cluster centroids and projects [180]
cells to the MST backbone to build the pseudotime course
Diffusion Reduces dimensionality with diffusion maps, uses weighted [182]
Pseudotime kNN graph and orders cells using a random-​walk-​based
(DPT) distance
FateID Uses random forests to quantify fate bias, which is then used to [191]
pseudo-​temporally order cells
Waterfall Builds MST on cell clusters, and use distance to cluster centers [181]
for orthogonal projection
Wanderlust Selects the shortest path from an ensemble of kNN graphs, [192]
and order cells along the path based on their distance to user-​
defined start cell and random waypoint cells
Wishbone An extension of Wanderlust for bifurcation branching topology [193]

classified as linear, bifurcating, multifurcating, cyclic, tree-​like, etc. While


some methods (e.g., TSCAN, Wanderlust, and Waterfall) are designed to infer
linear trajectories, many methods can infer multiple topologies, from linear,
bifurcating, multifurcating, to even tree-​like trajectories. Methods like PAGA
can infer even more complicated topologies such as cyclical, connected, or
disconnected graphs. Some of the methods, such as FateID, require prior
information from user input in the form of specifying a starting or ending
cell, or the number of branches, while others such as PAGA, Wanderlust, and
Wishbone can take advantage of such information, but this is not mandatory.
Besides using static snapshots of cellular states at a certain time point,
some methods are developed to infer trajectories from time series data, the
examples of which are TASIC [183], Waddington-​OT [184], CSHMM [185],
and Tempora [186]. Given the wide diversity of currently available TI tools,
benchmarking studies may help guide their selection. One comprehensive
evaluation of 45 currently available TI methods shows that (1) no one method
applies to all scenarios, (2) there is considerable complementarity between the
184 Next-Generation Sequencing Data Analysis

tested methods, and (3) the user should choose and use a variety of methods
based on the expected trajectory topology [178]. Based on this comparison,
the top performing methods include PAGA, Slingshot, different versions of
Monocle, as well as generic methods such as MST.
After inference of cellular trajectory, the next questions to ask are what
genes are associated with cell lineage development and what key genes
underlie transitions between cellular states. As indicated in the last section,
methods for DE gene analysis for trajectories are still limited. Besides
Monocle 3 and tradeSeq as mentioned in the last section, other available tra-
jectory DE methods include those employed by TSCAN, GPfates [187], and
earlier versions of Monocle. Monocle 1 uses generalized additive models
to test whether genes significantly change their expression as a function of
pseudotime. TSCAN employs a similar approach. Monocle 2 uses a different
approach called BEAM (branch expression analysis modeling) to test whether
gene expression changes are associated with cell lineage branching along a
trajectory. GPfates models gene expression-​dependent cell fates as temporal
mixtures of Gaussian processes. Similar to BEAM, it can identify gene expres-
sion changes associated with bifurcation points. Besides these tools that per-
form both trajectory inference and DE analysis, there are also tools that take
as input pseudotemporal ordering of cells inferred by the TI tools detailed
above, to conduct time-​course DE analysis. LineagePulse, representing such
an example, fits ZINB noise model to gene expression data collected from
pseudotemporally ordered single cells [188].
Using static snapshots of cellular states, TI does not make predictions
on the speed or direction of cell progression along the trajectory. To make
such predictions, additional information is required. Change in cellular
mRNA abundance inferred from the same static snapshots by a strategy
called RNA velocity analysis [189] provides such information. The RNA
velocity strategy is based on the detection and comparison of unspliced
pre-​mature transcripts that still contain introns, and spliced mature
transcripts. In principle, this strategy is built on the premise that if there is
a high ratio of unspliced to spliced mRNA molecules (called positive vel-
ocity) from a gene, it indicates that expression of the gene is upregulated
from its steady state. Conversely, if the ratio of unspliced to spliced mRNA
abundance is lower than its steady state ratio (i.e., negative velocity), it is
indicative of downregulation for the gene. Based on aggregation of RNA
velocities inferred across genes, this analysis then makes predictions on
the future state of each cell in terms of the speed and direction of their
movement along the trajectory. Currently available RNA velocity analysis
tools include VeloCyto [189] and scVelo [190]. These RNA velocity tools are
compatible and can be deployed with pipeline toolkits such as Seurat and
Scanpy. Because it adds predictive information onto a trajectory about the
direction and speed of cellular movement, RNA velocity analysis is often
carried out in combination with TI.
Transcriptomics by Single-Cell RNA-Seq 185

8.8 Advanced Analyses
8.8.1 SNV/​CNV Detection and Allele-​Specific Expression Analysis
Besides transcriptomic profiles, scRNA-​ seq data also contains genotypic
information specific for each cell, including single nucleotide and structural
variants. Such additional information is especially helpful for studies that
involve genome instability, such as cancer or other diseases related to aging.
The genotypic information embedded in scRNA-​seq data can help uncover
functional variants in individual cells, and may also inform their specific
gene expression pattern. Understandably, detection of these variants from
scRNA-​seq data is limited to expressed regions that have enough sequen-
cing depth. While some single-cell sequencing platforms such as Smart-​seq3
generate reads that cover the full length of transcripts, others such as 10×
Chromium focus on the 3’ or 5’ end of transcripts. RNA editing may also
add another layer of complication by revealing variants that may not be
present at the DNA level, but occurrence of RNA editing is typically very
rare. To detect SNVs, methods developed for calling variants from bulk
RNA-​seq data, such as MuTect2, Strelka2 [194], VarScan2, SAMtools [195],
Pysam [196], FreeBayes, and BamBam, can be used on scRNA-​seq data. The
GATK RNA-​ seq short variant discovery best practices workflow, which
uses HaplotyperCaller for variant calling followed by variant filtering using
RNA-​seq specific settings, is among the most used [197]. Monovar, a method
developed for single-cell DNA sequencing data [198], can also be used for
calling SNVs from scRNA-​seq data [199]. There are currently a number of
tools that have been developed for SNV detection from scRNA-​seq data,
including SSrGE [200], Trinity CTAT [201], and cellsnp-​lite [202]. Among
these methods, cellsnp-​lite is a lightweight allelic reads pileup method with
minimum filtering that can be applied to both 10× Chromium and Smart-​seq3
data. Because of use of parallel processing it has improved running speed.
Benchmarking comparison has shown that the performance of many current
tools depends on sequencing depth, genomic context (such as high GC con-
tent), functional region, variant allele frequency, and platform (10x has more
dropout events) [203]. It has also been shown that the main detection limi-
tation is low sensitivity caused by low capture efficiency, sequencing depth,
and signal dropout. Among the best performing tools so far are SAMtools,
FreeBayes, Strelka2, and CTAT.
Deletion or duplication of a genomic region may lead to reduced or increased
expression of genes located in the affected region. It is possible, therefore, to
infer CNV information from scRNA-​seq data. While it can be challenging
due to uneven coverage of scRNA-​seq signal across the genome, inferred
CNVs do provide information on cellular heterogeneity at another dimen-
sion (genome instability), for instance, during cancer development [204].
To meet the challenges of calling CNVs from scRNA-​seq data, a relatively
186 Next-Generation Sequencing Data Analysis

small number of methods, including HoneyBADGER [205], CaSpER [206],


inferCNV [207], and CONICS [208], are currently available. HoneyBADGER,
for example, uses a probabilistic hidden Markov model to infer CNV from
smoothed averaged gene expression profile of a set of cells in comparison to
control cells. CaSpER identifies and visualizes CNV events from single-cell
gene expression signals integrated with allelic shift signals, which quantifies
loss of heterozygosity events across the genome facilitating CNV detection.
InferCNV, a component of Trinity CTAT, searches for evidence of chromo-
somal segmental gains or losses by averaging expression over neighboring
genes across the genome, in comparison to reference normal cells. CONICS
(or CONICSmat when reference normal scRNA-​seq data is not available)
applies a two-​component Gaussian mixture model to fit gene expression in a
chromosomal region to make CNV calls.
To further connect genetic variants with gene expression regulation, allele-​
specific expression (ASE) analysis can be performed to determine potential
imbalanced expression of transcripts from each allele. Imbalanced or pref-
erential allelic expression leads to phenotypic variation, and is caused by
epigenetic or genetic regulatory mechanisms. ASE analysis has been widely
used to reveal allelic imbalance or allele-​specific transcriptional bursting from
bulk RNA-​seq. Single-cell ASE analysis is still relatively new, and as a result
available methods are still quite limited. Prashant et al. (2020) used a custom
pipeline to estimate ASE from 10× Chromium scRNA-​seq. This pipeline first
uses STAR to align raw reads pooled from all cells, and then GATK to call
all SNVs present in the data. Filtered high-​quality, heterozygous SNVs are
then used as input for a second round of STAR-​based alignment, in which an
SNV-​aware option such as WASP [209] is employed on reads within each cell.
To detect monoallelic and biallelic expression, VAFRNA, i.e., the percentage
of reads carrying the variant sequence, is then calculated. Besides directly
calling from scRNA-​seq data, the SNVs needed for the second round of
alignment and VAFRNA calculation can also be provided from bulk RNA-​seq
or DNA sequencing data [210]. Because of the inherent signal dropout issue
and potential allele-​mapping bias, interpretation of single-cell ASE results
should use caution.

8.8.2 Alternative Splicing Analysis


As mentioned in Chapter 7 on bulk RNA-​seq, alternative splicing plays an
important role in regulating major biological processes, and abnormal spli-
cing may lead to diseased states. At the single-cell level, alternative splicing
can be analyzed despite the challenges of signal sparsity and background
noise. Tools for this analysis include Outrigger [211], BRIE/​BRIE2 [212, 213],
ODEGR-​NMF [214], ASCOT [215], Millefy [216], VALERIE [217], SCATS [218],
scQuint [219], and DESJ-​detection [220]. To illustrate how these tools work,
Outrigger, a component of the Expedition suite, uses junction-​spanning reads
to create an exon-​junction graph first, and then detects alternative splicing
Transcriptomics by Single-Cell RNA-Seq 187

events through traversing the graph. Quantification of such events and iden-
tification of differential splicing between groups of cells are based on the
use of “percent-​spliced-​in” or Psi (ψ). As another example, DESJ-​detection
first constructs a cell-​splicing junction count matrix for each gene. Iterative
K-​means is then used to cluster cells; after removing clusters with low expres-
sion, a list of solid junctions is generated. The identification of DESJs, or dif-
ferentially expressed splicing junctions, is achieved using limma. To help
visualize differential splicing patterns across cells, Millefy and VALERIE can
be used to uncover cellular heterogeneity and splicing differences between
various cell groups. VALERIE, for example, is an R-​based tool for using Psi
values to display alternative splicing events. It can also be used to identify
significant splicing difference between different cell populations, through
performing statistical test, such as Kruskal–​Wallis test, on the Psi values
followed by multiple testing correction. Most of alternative splicing ana-
lysis tools use reads obtained from full-​length scRNA-​seq platforms such as
Smart-​seq3. Reads derived from the 3’ or 5’ end of transcripts, such as those
generated from the 10× Chromium platform, cover limited number of spli-
cing junctions at either end of genes. Despite this limitation, some tools such
as SCATS can use 10× scRNA-​seq data for alternative splicing analysis.

8.8.3 Gene Regulatory Network Inference


A gene regulatory network (GRN) is a graph representation of how genes
interact with each other in a cell system. In a GRN, genes are represented as
nodes, and their interactions constitute edges. Compared to the analytic steps
above, GRN inference is relatively new and yet to be performed more widely.
Currently there are a growing number of scRNA-​seq based GRN inference
methods available, including SCENIC [221], SCODE [222], SCOUP [223],
PIDC [224], LEAP [225], NLNET [226], SCIMITAR [227], and GRISLI [228].
In addition, some of the methods originally developed for bulk RNA-​seq
data have also been applied to scRNA-​seq data after some adjustment with
good performance, including GENIE3 [229] and GRNBoost2 [230]. GENIE3
is a random forest regression-​based algorithm that had the best perform-
ance in the DREAM4 In Silico Multifactorial challenge for assessment of GRN
inference algorithms. GRNBoost2, based on the use of a stochastic gradient
boosting machine regression model, has been developed as a faster alterna-
tive to GENIE3. These, and the methods specifically developed for scRNA-​
seq, use the general principle of gene co-​expression to infer GRN. If two
genes show co-​expression in the context of other genes, they are considered
to interact.
According to how they infer GRNs, the algorithms that are specific-
ally developed for scRNA-​seq data can be grouped into several categories.
Methods such as SCENIC, PIDC, and NLNET are based on simple correlation
analysis. SCENIC, for example, identifies sets of genes that are co-​expressed
188 Next-Generation Sequencing Data Analysis

with transcription factors with the use of GENIE3. To trim the large number
of edges that represent potential gene-​gene interactions to a shorter list of
high-​confidence edges, SCENIC performs a transcription factor-​ binding
motif enrichment analysis to identify putative target genes. The output from
SCENIC can then be imported into visualization tools such SCope for net-
work visualization. Methods such as LEAP and SCIMITAR take into con-
sideration effects of developmental stages on gene networking through
incorporating pseudo-​temporal information from trajectory inference and
velocity analysis (Section 8.7). The pseudo-​temporal ordering of cells helps
establish directionality between an upstream gene and a downstream effector.
For these methods, gene correlation is first calculated for each time window,
and the multiple correlation matrices are then aggregated into one adjacency
matrix to represent the overall gene-​gene interactions. Other methods such as
SCODE, SCOUP, and GRISLI use a similar approach with the application of
pseudo-​temporal information, but they use differential equations to estimate
gene correlation and infer gene relationships. For example, SCODE relies on
Monocle to provide pseudotime information, and uses ordinary differential
equations to calculate gene correlation. There are also methods based on other
approaches, such as SCNS [231] and BTR [232] that use Boolean models. With
such models 0 or 1 represents deactivated or activated gene expression, and
Boolean operations AND, OR, and NOT are used to capture relationships
between two genes. Boolean models provide a simplistic presentation of the
cell system through converting gene expression data into binary data, but
this also leads to loss of gene-​gene interaction information.
To pick an appropriate GRN inference method, besides having knowledge
of how they are designed, it also helps to understand whether prior infor-
mation is required and what is the basic characteristic of the cells under
study. Some of the methods require prior information, in the form of pseudo-​
temporal ordering of cells or cell types, which can be revealed with trajec-
tory inference. Such methods are more suitable for cells that are in different
developmental stages. If the objective is to compare cellular composition or
heterogeneity under different conditions (e.g., healthy vs. diseased), methods
that employ static data are more appropriate. To provide systematic guidance
to the selection of appropriate GRN inference methods, results from several
benchmark studies [233–​236] are available on currently available GRN infer-
ence methods. Overall these studies showed underperformance of current
methods and call for development of better designed tools. For example, cur-
rently inferred networks still show poor agreement with ground truth. In the
meantime, these studies also revealed the challenges of inferring GRN from
scRNA-​seq data, which can be technical (due mostly to signal heterogeneity
and sparsity), biological (e.g., complex nature of molecular interactions), or
computational (e.g., complexity of analysis). Validation of an inferred GRN
is also a very challenging task. Most of the currently available methods still
Transcriptomics by Single-Cell RNA-Seq 189

output one gene network for all cells, or for specific cell types, not individual
cells. Some newer methods such as CSN [237] and c-​CSN [238] allow building
of cell-​specific networks, i.e., one network per cell.
Despite the current challenges, the value of GRN analysis cannot be
overemphasized. In-​depth analysis of a GRN enables detection of network
modules and key nodes (hub genes). A module refers to a group of genes
that are highly connected to fulfill a cellular function. The overall topology
of a module, or key nodes linking different modules, might change with
development or cell differentiation, or differ under different conditions.
Differential network analysis may reveal altered gene-​ gene interactions
between conditions. These network analyses can be carried out using tools
such as WGCNA [239].

References
1. Cui Y, Irudayaraj J. Inside single cells: quantitative analysis with advanced
optics and nanomaterials. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015,
7(3):387–​407.
2. Huang XT, Li X, Qin PZ, Zhu Y, Xu SN, Chen JP. Technical advances in single-​
cell RNA sequencing and applications in normal and malignant hematopoi-
esis. Front Oncol 2018, 8:582.
3. Shalek AK, Satija R, Shuga J, Trombetta JJ, Gennert D, Lu D, Chen P, Gertner
RS, Gaublomme JT, Yosef N et al. Single-​cell RNA-​seq reveals dynamic para-
crine control of cellular variation. Nature 2014, 510(7505):363–​369.
4. Marinov GK, Williams BA, McCue K, Schroth GP, Gertz J, Myers RM, Wold BJ.
From single-​cell to cell-​pool transcriptomes: stochasticity in gene expression
and RNA splicing. Genome Res 2014, 24(3):496–​510.
5. Zhang M, Zou Y, Xu X, Zhang X, Gao M, Song J, Huang P, Chen Q, Zhu Z, Lin
W et al. Highly parallel and efficient single cell mRNA sequencing with paired
picoliter chambers. Nat Commun 2020, 11(1):2118.
6. Hagemann-​ Jensen M, Ziegenhain C, Chen P, Ramskold D, Hendriks GJ,
Larsson AJM, Faridani OR, Sandberg R. Single-​cell RNA counting at allele and
isoform resolution using Smart-​seq3. Nat Biotechnol 2020, 38(6):708–​714.
7. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I,
Bialas AR, Kamitaki N, Martersteck EM et al. Highly Parallel Genome-​wide
Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell 2015,
161(5):1202–​1214.
8. Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L,
Weitz DA, Kirschner MW. Droplet barcoding for single-​cell transcriptomics
applied to embryonic stem cells. Cell 2015, 161(5):1187–​1201.
9. Cao J, Packer JS, Ramani V, Cusanovich DA, Huynh C, Daza R, Qiu X, Lee C,
Furlan SN, Steemers FJ et al. Comprehensive single-​cell transcriptional pro-
filing of a multicellular organism. Science 2017, 357(6352):661–​667.
190 Next-Generation Sequencing Data Analysis

10. Zheng GX, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB,
Wheeler TD, McDermott GP, Zhu J et al. Massively parallel digital transcrip-
tional profiling of single cells. Nat Commun 2017, 8:14049.
11. Zhang X, Li T, Liu F, Chen Y, Yao J, Li Z, Huang Y, Wang J. Comparative ana-
lysis of droplet-​based ultra-​high-​throughput single-​cell RNA-​seq systems.
Mol Cell 2019, 73(1):130–​142 e135.
12. Ding J, Adiconis X, Simmons SK, Kowalczyk MS, Hession CC, Marjanovic
ND, Hughes TK, Wadsworth MH, Burks T, Nguyen LT et al. Systematic com-
parison of single-​ cell and single-​ nucleus RNA-​ sequencing methods. Nat
Biotechnol 2020, 38(6):737–​746.
13. How many Cells (https://​satija​lab.org/​howma​nyce​lls)
14. Svensson V, da Veiga Beltrame E, Pachter L. Quantifying the tradeoff between
sequencing depth and cell number in single-​ cell RNA-​ seq. bioRxiv 2019,
doi: https://​doi.org/​10.1101/​762​773
15. Zhang MJ, Ntranos V, Tse D. Determining sequencing depth in a single-​cell
RNA-​seq experiment. Nat Commun 2020, 11(1):774.
16. Parekh S, Ziegenhain C, Vieth B, Enard W, Hellmann I. zUMIs –​A fast and
flexible pipeline to process RNA sequencing data with UMIs. GigaScience 2018,
7(6):giy059.
17. Heimberg G, Bhatnagar R, El-​Samad H, Thomson M. Low dimensionality
in gene expression data enables the accurate extraction of transcriptional
programs from shallow sequencing. Cell Syst 2016, 2(4):239–​250.
18. Wu AR, Neff NF, Kalisky T, Dalerba P, Treutlein B, Rothenberg ME, Mburu
FM, Mantalas GL, Sim S, Clarke MF et al. Quantitative assessment of single-​
cell RNA-​sequencing methods. Nat Methods 2014, 11(1):41–​46.
19. Svensson V, Natarajan KN, Ly LH, Miragaia RJ, Labalette C, Macaulay IC,
Cvejic A, Teichmann SA. Power analysis of single-​ cell RNA-​ sequencing
experiments. Nat Methods 2017, 14(4):381–​387.
20. Genomics X. Technical Note–​ Removal of Dead Cells from Single Cell
Suspensions Improves Performance for 10× Genomics® Single Cell
Applications. 2017.
21. van den Brink SC, Sage F, Vertesy A, Spanjaard B, Peterson-​Maduro J, Baron
CS, Robin C, van Oudenaarden A. Single-​cell sequencing reveals dissociation-​
induced gene expression in tissue subpopulations. Nat Methods 2017,
14(10):935–​936.
22. Wohnhaas CT, Leparc GG, Fernandez-​Albert F, Kind D, Gantner F, Viollet
C, Hildebrandt T, Baum P. DMSO cryopreservation is the method of choice
to preserve cells for droplet-​based single-​cell RNA sequencing. Sci Rep 2019,
9(1):10699.
23. Cha J, Lee I. Single-​cell network biology for resolving cellular heterogeneity in
human diseases. Exp Mol Med 2020, 52(11):1798–​1808.
24. Korrapati S, Taukulis I, Olszewski R, Pyle M, Gu S, Singh R, Griffiths C, Martin
D, Boger E, Morell RJ et al. Single cell and single nucleus RNA-​seq reveal cel-
lular heterogeneity and homeostatic regulatory networks in adult mouse stria
vascularis. Front Mol Neurosci 2019, 12:316.
25. Gao R, Kim C, Sei E, Foukakis T, Crosetto N, Chan LK, Srinivasan M, Zhang
H, Meric-​Bernstam F, Navin N. Nanogrid single-​nucleus RNA sequencing
reveals phenotypic diversity in breast cancer. Nat Commun 2017, 8(1):228.
Transcriptomics by Single-Cell RNA-Seq 191

26. Liang Q, Dharmat R, Owen L, Shakoor A, Li Y, Kim S, Vitale A, Kim I, Morgan


D, Liang S et al. Single-​nuclei RNA-​seq on human retinal tissue provides
improved transcriptome profiling. Nat Commun 2019, 10(1):5743.
27. Zhu YY, Machleder EM, Chenchik A, Li R, Siebert PD. Reverse transcriptase
template switching: a SMART approach for full-​length cDNA library con-
struction. BioTechniques 2001, 30(4):892–​897.
28. Bray NL, Pimentel H, Melsted P, Pachter L. Near-​optimal probabilistic RNA-​
seq quantification. Nat Biotechnol 2016, 34(5):525–​527.
29. Du Y, Huang Q, Arisdakessian C, Garmire LX. Evaluation of STAR and Kallisto
on single cell RNA-​seq data alignment. G3 2020, 10(5):1775–​1783.
30. Melsted P, Ntranos V, Pachter L. The barcode, UMI, set format and BUStools.
Bioinformatics 2019, 35(21):4472–​4473.
31. Srivastava A, Malik L, Smith T, Sudbery I, Patro R. Alevin efficiently estimates
accurate gene abundances from dscRNA-​seq data. Genome Biol 2019, 20(1):65.
32. McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: doublet detection in
single-​cell RNA sequencing data using artificial nearest neighbors. Cell Syst
2019, 8(4):329–​337 e324.
33. Wolock SL, Lopez R, Klein AM. Scrublet: Computational identification of cell
doublets in single-​cell transcriptomic data. Cell Syst 2019, 8(4):281–​291 e289.
34. Gayoso A, Shor J, Carr AJ, Sharma R, Pe’er D. DoubletDetection. In.: Zenodo;
2020: https://​zen​odo.org/​rec​ord/​2678​042.
35. Bais AS, Kostka D. scds: computational annotation of doublets in single-​cell
RNA sequencing data. Bioinformatics 2020, 36(4):1150–​1158.
36. Bernstein NJ, Fong NL, Lam I, Roy MA, Hendrickson DG, Kelley DR.
Solo: doublet identification in single-​cell RNA-​seq via semi-​supervised deep
learning. Cell Syst 2020, 11(1):95–​101 e105.
37. Xi NM, Li JJ. Benchmarking computational doublet-​detection methods for
single-​cell RNA sequencing data. Cell Syst 2020, 12(2):176–​194.
38. Stoeckius M, Zheng S, Houck-​Loomis B, Hao S, Yeung BZ, Mauck WM,
3rd, Smibert P, Satija R. Cell Hashing with barcoded antibodies enables
multiplexing and doublet detection for single cell genomics. Genome Biol 2018,
19(1):224.
39. Kang HM, Subramaniam M, Targ S, Nguyen M, Maliskova L, McCarthy E,
Wan E, Wong S, Byrnes L, Lanata CM et al. Multiplexed droplet single-​cell
RNA-​sequencing using natural genetic variation. Nat Biotechnol 2018, 36(1):89.
40. Xu J, Falconer C, Nguyen Q, Crawford J, McKinnon BD, Mortlock S, Senabouth
A, Andersen S, Chiu HS, Jiang LD et al. Genotype-​free demultiplexing of
pooled single-​cell RNA-​seq. Genome Biol 2019, 20(1):290.
41. Heaton H, Talman AM, Knights A, Imaz M, Gaffney DJ, Durbin R, Hemberg
M, Lawniczak MKN. Souporcell: robust clustering of single-​cell RNA-​seq data
by genotype without reference genotypes. Nat Methods 2020, 17(6):615.
42. Huang YH, McCarthy DJ, Stegle O. Vireo: Bayesian demultiplexing of pooled
single-​cell RNA-​ seq data without genotype reference. Genome Biol 2019,
20(1):273.
43. Lun ATL, Riesenfeld S, Andrews T, Dao TP, Gomes T, participants in the 1st
Human Cell Atlas J, Marioni JC. EmptyDrops: distinguishing cells from empty
droplets in droplet-​based single-​cell RNA sequencing data. Genome Biol 2019,
20(1):63.
192 Next-Generation Sequencing Data Analysis

44. Heiser CN, Wang VM, Chen B, Hughey JJ, Lau KS. Automated quality con-
trol and cell identification of droplet-​based single-​cell data using dropkick.
Genome Res 2021, 31(10):1742–​1752 .
45. Ni Z, Chen S, Brown J, Kendziorski C. CB2 improves power of cell detec-
tion in droplet-​based single-​cell RNA sequencing data. Genome Biol 2020,
21(1):137.
46. Yang S, Corbett SE, Koga Y, Wang Z, Johnson WE, Yajima M, Campbell JD.
Decontamination of ambient RNA in single-​ cell RNA-​seq with DecontX.
Genome Biol 2020, 21(1):57.
47. Young MD, Behjati S. SoupX removes ambient RNA contamination from
droplet-​based single-​cell RNA sequencing data. Gigascience 2020, 9(12):giaa151.
48. Osorio D, Cai JJ. Systematic determination of the mitochondrial proportion in
human and mice tissues for single-​cell RNA sequencing data quality control.
Bioinformatics 2020, 37(7):963–​967.
49. Bacher R, Chu LF, Leng N, Gasch AP, Thomson JA, Stewart RM, Newton M,
Kendziorski C. SCnorm: robust normalization of single-​cell RNA-​seq data.
Nat Methods 2017, 14(6):584–​586.
50. Yip SH, Wang P, Kocher JA, Sham PC, Wang J. Linnorm: improved statis-
tical analysis for single cell RNA-​seq expression data. Nucleic Acids Res 2017,
45(22):e179.
51. Vallejos CA, Marioni JC, Richardson S. BASiCS: Bayesian Analysis of Single-​
Cell Sequencing Data. PLoS Comput Biol 2015, 11(6):e1004333.
52. Qiu X, Hill A, Packer J, Lin D, Ma YA, Trapnell C. Single-​cell mRNA
quantification and differential analysis with Census. Nat Methods 2017,
14(3):309–​315.
53. Risso D, Perraudeau F, Gribkova S, Dudoit S, Vert JP. A general and flexible
method for signal extraction from single-​cell RNA-​seq data. Nat Commun
2018, 9(1):284.
54. Hafemeister C, Satija R. Normalization and variance stabilization of single-​
cell RNA-​seq data using regularized negative binomial regression. Genome
Biol 2019, 20(1):296.
55. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-​cell
transcriptomic data across different conditions, technologies, and species. Nat
Biotechnol 2018, 36(5):411–​420.
56. Wolf FA, Angerer P, Theis FJ. SCANPY: large-​scale single-​cell gene expression
data analysis. Genome Biol 2018, 19(1):15.
57. Lun AT, Bach K, Marioni JC. Pooling across cells to normalize single-​cell RNA
sequencing data with many zero counts. Genome Biol 2016, 17:75.
58. Lopez R, Regier J, Cole MB, Jordan MI, Yosef N. Deep generative modeling for
single-​cell transcriptomics. Nat Methods 2018, 15(12):1053–​1058.
59. Lytal N, Ran D, An L. Normalization methods on single-​cell RNA-​seq data: an
empirical survey. Front Genet 2020, 11:41.
60. Cole MB, Risso D, Wagner A, DeTomaso D, Ngai J, Purdom E, Dudoit S, Yosef
N. Performance Assessment and Selection of Normalization Procedures for
Single-​Cell RNA-​Seq. Cell Syst 2019, 8(4):315–​328 e318.
61. Buttner M, Miao Z, Wolf FA, Teichmann SA, Theis FJ. A test metric for assessing
single-​cell RNA-​seq batch correction. Nat Methods 2019, 16(1):43–​49.
62. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expres-
sion data using empirical Bayes methods. Biostatistics 2007, 8(1):118–​127.
Transcriptomics by Single-Cell RNA-Seq 193

63. Haghverdi L, Lun ATL, Morgan MD, Marioni JC. Batch effects in single-​cell
RNA-​sequencing data are corrected by matching mutual nearest neighbors.
Nat Biotechnol 2018, 36(5):421–​427.
64. Stuart T, Satija R. Integrative single-​ cell analysis. Nat Rev Genet 2019,
20(5):257–​272.
65. Welch JD, Kozareva V, Ferreira A, Vanderburg C, Martin C, Macosko EZ.
Single-​cell multi-​omic integration compares and contrasts features of brain
cell identity. Cell 2019, 177(7):1873–​1887 e1817.
66. Hie B, Bryson B, Berger B. Efficient integration of heterogeneous single-​cell
transcriptomes using Scanorama. Nat Biotechnol 2019, 37(6):685–​691.
67. Polanski K, Young MD, Miao Z, Meyer KB, Teichmann SA, Park JE.
BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics
2020, 36(3):964–​965.
68. Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y,
Brenner M, Loh PR, Raychaudhuri S. Fast, sensitive and accurate integration
of single-​cell data with Harmony. Nat Methods 2019, 16(12):1289–​1296.
69. Barkas N, Petukhov V, Nikolaeva D, Lozinsky Y, Demharter S, Khodosevich K,
Kharchenko PV. Joint analysis of heterogeneous single-​cell RNA-​seq dataset
collections. Nat Methods 2019, 16(8):695–​698.
70. Tran HTN, Ang KS, Chevrier M, Zhang X, Lee NYS, Goh M, Chen J. A bench-
mark of batch-​effect correction methods for single-​cell RNA sequencing data.
Genome Biol 2020, 21(1):12.
71. Rousseeuw PJ. Silhouettes: a graphical aid to the interpretation and validation
of cluster analysis. J Comput Appl Math 1987, 20:53–​65.
72. van Dijk D, Sharma R, Nainys J, Yim K, Kathail P, Carr AJ, Burdziak C, Moon
KR, Chaffer CL, Pattabiraman D et al. Recovering gene interactions from
single-​cell data using data diffusion. Cell 2018, 174(3):716–​729 e727.
73. Wagner F, Yan Y, Yanai I. K-​nearest neighbor smoothing for high-​throughput
single-​cell RNA-​Seq data. bioRxiv 2018, doi: https://​doi.org/​10.1101/​217​737
74. Huang M, Wang J, Torre E, Dueck H, Shaffer S, Bonasio R, Murray JI, Raj A, Li
M, Zhang NR. SAVER: gene expression recovery for single-​cell RNA sequen-
cing. Nat Methods 2018, 15(7):539–​542.
75. Wang J, Agarwal D, Huang M, Hu G, Zhou Z, Ye C, Zhang NR. Data denoising
with transfer learning in single-​ cell transcriptomics. Nat Methods 2019,
16(9):875–​878.
76. Linderman GC, Zhao J, Roulis M, Bielecki P, Flavell RA, Nadler B, Kluger Y.
Zero-​preserving imputation of single-​cell RNA-​seq data. Nat Commun 2022,
13(1):192.
77. Lin PJ, Troup M, Ho JWK. CIDR: Ultrafast and accurate clustering through
imputation for single-​cell RNA-​seq data. Genome Biol 2017, 18(1):59.
78. Eraslan G, Simon LM, Mircea M, Mueller NS, Theis FJ. Single-​cell RNA-​seq
denoising using a deep count autoencoder. Nat Commun 2019, 10(1):390.
79. Li WV, Li JJ. An accurate and robust imputation method scImpute for single-​
cell RNA-​seq data. Nat Commun 2018, 9(1):997.
80. Mongia A, Sengupta D, Majumdar A. McImpute: matrix completion based
imputation for single cell RNA-​seq data. Front Genet 2019, 10:9.
81. Gong W, Kwak IY, Pota P, Koyano-​ Nakagawa N, Garry DJ. DrImpute:
imputing dropout events in single cell RNA sequencing data. BMC
Bioinformatics 2018, 19(1):220.
194 Next-Generation Sequencing Data Analysis

82. Lahnemann D, Koster J, Szczurek E, McCarthy DJ, Hicks SC, Robinson MD,
Vallejos CA, Campbell KR, Beerenwinkel N, Mahfouz A et al. Eleven grand
challenges in single-​cell data science. Genome Biol 2020, 21(1):31.
83. Andrews TS, Hemberg M. False signals induced by single-​cell imputation.
F1000Research 2018, 7:1740.
84. Hou W, Ji Z, Ji H, Hicks SC. A systematic evaluation of single-​cell RNA-​
sequencing imputation methods. Genome Biol 2020, 21(1):218.
85. Li Y, Willer C, Sanna S, Abecasis G. Genotype imputation. Annu Rev Genomics
Hum Genet 2009, 10:387–​406.
86. Brennecke P, Anders S, Kim JK, Kolodziejczyk AA, Zhang X, Proserpio V,
Baying B, Benes V, Teichmann SA, Marioni JC et al. Accounting for technical
noise in single-​cell RNA-​seq experiments. Nat Methods 2013, 10(11):1093–​1095.
87. Buettner F, Natarajan KN, Casale FP, Proserpio V, Scialdone A, Theis FJ,
Teichmann SA, Marioni JC, Stegle O. Computational analysis of cell-​to-​cell het-
erogeneity in single-​cell RNA-​sequencing data reveals hidden subpopulations
of cells. Nat Biotechnol 2015, 33(2):155–​160.
88. Yip SH, Sham PC, Wang J. Evaluation of tools for highly variable gene dis-
covery from single-​cell RNA-​seq data. Brief Bioinform 2019, 20(4):1583–​1589.
89. Duo A, Robinson MD, Soneson C. A systematic performance evaluation of
clustering methods for single-​cell RNA-​seq data. F1000Research 2018, 7:1141.
90. Townes FW, Hicks SC, Aryee MJ, Irizarry RA. Feature selection and dimen-
sion reduction for single-​cell RNA-​Seq based on a multinomial model. Genome
Biol 2019, 20(1):295.
91. Andrews TS, Hemberg M. M3Drop: dropout-​ based feature selection for
scRNASeq. Bioinformatics 2019, 35(16):2865–​2867.
92. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon
NJ, Livak KJ, Mikkelsen TS, Rinn JL. The dynamics and regulators of cell
fate decisions are revealed by pseudotemporal ordering of single cells. Nat
Biotechnol 2014, 32(4):381–​386.
93. Shao C, Hofer T. Robust classification of single-​cell transcriptome data by non-
negative matrix factorization. Bioinformatics 2017, 33(2):235–​242.
94. Pierson E, Yau C. ZIFA: Dimensionality reduction for zero-​inflated single-​cell
gene expression analysis. Genome Biol 2015, 16:241.
95. Buettner F, Pratanwanich N, McCarthy DJ, Marioni JC, Stegle O. f-​scLVM: scal-
able and versatile factor analysis for single-​cell RNA-​seq. Genome Biol 2017,
18(1):212.
96. Mahfouz A, van de Giessen M, van der Maaten L, Huisman S, Reinders M,
Hawrylycz MJ, Lelieveldt BP. Visualizing the spatial gene expression organ-
ization in the brain through non-​linear similarity embeddings. Methods 2015,
73:79–​89.
97. Senabouth A, Lukowski SW, Hernandez JA, Andersen SB, Mei X, Nguyen
QH, Powell JE. ascend: R package for analysis of single-​cell RNA-​seq data.
GigaScience 2019, 8(8):giz087.
98. Sun S, Zhu J, Ma Y, Zhou X. Accuracy, robustness and scalability of
dimensionality reduction methods for single-​cell RNA-​seq analysis. Genome
Biol 2019, 20(1):269.
99. Welch JD, Hartemink AJ, Prins JF. SLICER: inferring branched, nonlinear cel-
lular trajectories from single cell RNA-​seq data. Genome Biol 2016, 17(1):106.
Transcriptomics by Single-Cell RNA-Seq 195

100. Haghverdi L, Buettner F, Theis FJ. Diffusion maps for high-​dimensional single-​
cell analysis of differentiation data. Bioinformatics 2015, 31(18):2989–​2998.
101. Sun X, Liu Y, An L. Ensemble dimensionality reduction and feature gene
extraction for single-​cell RNA-​seq data. Nat Commun 2020, 11(1):5853.
102. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, Ginhoux F,
Newell EW. Dimensionality reduction for visualizing single-​cell data using
UMAP. Nat Biotechnol 2018, 37(1):38–​44.
103. Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, Hill AJ, Zhang F, Mundlos
S, Christiansen L, Steemers FJ et al. The single-​cell transcriptional landscape
of mammalian organogenesis. Nature 2019, 566(7745):496–​502.
104. Kobak D, Berens P. The art of using t-​SNE for single-​cell transcriptomics. Nat
Commun 2019, 10(1):5416.
105. Hu Q, Greene CS. Parameter tuning is a key part of dimensionality reduction
via deep variational autoencoders for single cell RNA transcriptomics. Pac
Symp Biocomput 2019, 24:362–​373.
106. Ding J, Condon A, Shah SP. Interpretable dimensionality reduction of single
cell transcriptome data with deep generative models. Nat Commun 2018,
9(1):2002.
107. Deng Y, Bao F, Dai QH, Wu LF, Altschuler SJ. Scalable analysis of cell-​type
composition from single-​cell transcriptomics using deep recurrent learning.
Nature Methods 2019, 16(4):311.
108. Wang B, Zhu J, Pierson E, Ramazzotti D, Batzoglou S. Visualization and ana-
lysis of single-​cell RNA-​seq data by kernel-​based similarity learning. Nat
Methods 2017, 14(4):414–​416.
109. Xiang R, Wang W, Yang L, Wang S, Xu C, Chen X. A comparison for
dimensionality reduction methods of single-​cell RNA-​seq data. Front Genet
2021, 12:646936.
110. Wolf FA, Hamey FK, Plass M, Solana J, Dahlin JS, Gottgens B, Rajewsky N,
Simon L, Theis FJ. PAGA: graph abstraction reconciles clustering with trajec-
tory inference through a topology preserving map of single cells. Genome Biol
2019, 20(1):59.
111. Moon KR, van Dijk D, Wang Z, Gigante S, Burkhardt DB, Chen WS, Yim K,
Elzen AVD, Hirn MJ, Coifman RR et al. Visualizing structure and transitions
in high-​dimensional biological data. Nat Biotechnol 2019, 37(12):1482–​1492.
112. Anchang B, Hart TD, Bendall SC, Qiu P, Bjornson Z, Linderman M, Nolan
GP, Plevritis SK. Visualization and cellular hierarchy inference of single-​cell
data using SPADE. Nat Protoc 2016, 11(7):1264–​1279.
113. Weinreb C, Wolock S, Klein AM. SPRING: a kinetic interface for visual-
izing high dimensional single-​ cell expression data. Bioinformatics 2018,
34(7):1246–​1248.
114. Kim T, Chen IR, Lin Y, Wang AY, Yang JYH, Yang P. Impact of similarity
metrics on single-​ cell RNA-​ seq data clustering. Brief Bioinform 2019,
20(6):2316–​2326.
115. Moussa M, Mandoiu, II. Single cell RNA-​seq data clustering using TF-​IDF
based methods. BMC Genomics 2018, 19(Suppl 6):569.
116. Blondel VD, Guillaume JL, Lambiotte R, Lefebvre E. Fast unfolding of com-
munities in large networks. J Stat Mech-​Theory E 2008, doi:10.1088/​1742-​
5468/​2008/​10/​P10008
196 Next-Generation Sequencing Data Analysis

117. Traag VA, Waltman L, van Eck NJ. From Louvain to Leiden: guaranteeing
well-​connected communities. Sci Rep 2019, 9(1):5233.
118. Kiselev VY, Kirschner K, Schaub MT, Andrews T, Yiu A, Chandra T, Natarajan
KN, Reik W, Barahona M, Green AR et al. SC3: consensus clustering of single-​
cell RNA-​seq data. Nat Methods 2017, 14(5):483–​486.
119. Zurauskiene J, Yau C. pcaReduce: hierarchical clustering of single cell tran-
scriptional profiles. BMC Bioinformatics 2016, 17:140.
120. Grun D, Lyubimova A, Kester L, Wiebrands K, Basak O, Sasaki N, Clevers
H, van Oudenaarden A. Single-​cell messenger RNA sequencing reveals rare
intestinal cell types. Nature 2015, 525(7568):251–​255.
121. Xu C, Su Z. Identification of cell types from single-​cell transcriptomes using
a novel clustering method. Bioinformatics 2015, 31(12):1974–​1980.
122. Lake BB, Chen S, Sos BC, Fan J, Kaeser GE, Yung YC, Duong TE, Gao D,
Chun J, Kharchenko PV et al. Integrative single-​cell analysis of transcrip-
tional and epigenetic states in the human adult brain. Nat Biotechnol 2018,
36(1):70–​80.
123. Guo M, Wang H, Potter SS, Whitsett JA, Xu Y. SINCERA: a pipeline for single-​
cell RNA-​seq profiling analysis. PLoS Comput Biol 2015, 11(11):e1004575.
124. Freytag S, Tian L, Lonnstedt I, Ng M, Bahlo M. Comparison of clustering
tools in R for medium-​sized 10× Genomics single-​cell RNA-​sequencing data.
F1000Research 2018, 7:1297.
125. Yu D, Huber W, Vitek O. Shrinkage estimation of dispersion in negative bino-
mial models for RNA-​seq experiments with small sample size. Bioinformatics
2013, 29(10):1275–​1282.
126. Pliner HA, Shendure J, Trapnell C. Supervised classification enables rapid
annotation of cell atlases. Nat Methods 2019, 16(10):983–​986.
127. Domanskyi S, Szedlak A, Hawkins NT, Wang J, Paternostro G, Piermarocchi
C. Polled Digital Cell Sorter (p-​DCS): Automatic identification of hema-
tological cell types from single cell RNA-​ sequencing clusters. BMC
Bioinformatics 2019, 20(1):369.
128. Zhang Z, Luo D, Zhong X, Choi JH, Ma Y, Wang S, Mahrt E, Guo W, Stawiski
EW, Modrusan Z et al. SCINA: A semi-​supervised subtyping algorithm of
single cells and bulk samples. Genes 2019, 10(7):531.
129. Zhang AW, O’Flanagan C, Chavez EA, Lim JLP, Ceglia N, McPherson
A, Wiens M, Walters P, Chan T, Hewitson B et al. Probabilistic cell-​type
assignment of single-​cell RNA-​seq for tumor microenvironment profiling.
Nat Methods 2019, 16(10):1007–​1015.
130. Xu C, Lopez R, Mehlman E, Regier J, Jordan MI, Yosef N. Probabilistic har-
monization and annotation of single-​cell transcriptomics data with deep
generative models. Mol Syst Biol 2021, 17(1):e9620.
131. Franzen O, Gan LM, Bjorkegren JLM. PanglaoDB: a web server for explor-
ation of mouse and human single-​ cell RNA sequencing data. Database
(Oxford) 2019, 2019.
132. Zhang X, Lan Y, Xu J, Quan F, Zhao E, Deng C, Luo T, Xu L, Liao G, Yan M
et al. CellMarker: a manually curated resource of cell markers in human and
mouse. Nucleic Acids Res 2019, 47(D1):D721–​D728.
133. Zeisel A, Hochgerner H, Lonnerberg P, Johnsson A, Memic F, van der Zwan
J, Haring M, Braun E, Borm LE, La Manno G et al. Molecular architecture of
the mouse nervous system. Cell 2018, 174(4):999–​1014 e1022.
Transcriptomics by Single-Cell RNA-Seq 197

134. Ecker JR, Geschwind DH, Kriegstein AR, Ngai J, Osten P, Polioudakis D,
Regev A, Sestan N, Wickersham IR, Zeng H. The BRAIN Initiative Cell
Census Consortium: Lessons Learned toward Generating a Comprehensive
Brain Cell Atlas. Neuron 2017, 96(3):542–​557.
135. Saunders A, Macosko EZ, Wysoker A, Goldman M, Krienen FM, de
Rivera H, Bien E, Baum M, Bortolin L, Wang S et al. Molecular diversity
and specializations among the cells of the adult mouse brain. Cell 2018,
174(4):1015–​1030 e1016.
136. Regev A, Teichmann SA, Lander ES, Amit I, Benoist C, Birney E, Bodenmiller
B, Campbell P, Carninci P, Clatworthy M et al. The Human Cell Atlas. Elife
2017, 6:e27041.
137. Tabula Muris C, Overall c, Logistical c, Organ c, processing, Library p,
sequencing, Computational data a, Cell type a, Writing g et al. Single-​cell
transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 2018,
562(7727):367–​372.
138. Kiselev VY, Yiu A, Hemberg M. scmap: projection of single-​cell RNA-​seq
data across data sets. Nat Methods 2018, 15(5):359–​362.
139. Tan Y, Cahan P. SingleCellNet: a computational tool to classify single cell
RNA-​seq data across platforms and across species. Cell Syst 2019, 9(2):207–​
213 e202.
140. Ma F, Pellegrini M. ACTINN: automated identification of cell types in single
cell RNA sequencing. Bioinformatics 2020, 36(2):533–​538.
141. Alquicira-​Hernandez J, Sathe A, Ji HP, Nguyen Q, Powell JE. scPred: accurate
supervised method for cell-​type classification from single-​cell RNA-​seq data.
Genome Biol 2019, 20(1):264.
142. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, Blondel
M, Prettenhofer P, Weiss R, Dubourg V. Scikit-​learn: Machine learning in
Python. J Mach Learn Res 2011, 12:2825–​2830.
143. Bakken T, Cowell L, Aevermann BD, Novotny M, Hodge R, Miller JA, Lee A,
Chang I, McCorrison J, Pulendran B et al. Cell type discovery and represen-
tation in the era of high-​content single cell phenotyping. BMC Bioinformatics
2017, 18(Suppl 17):559.
144. Wang S, Pisco AO, McGeever A, Brbic M, Zitnik M, Darmanis S, Leskovec J,
Karkanias J, Altman RB. Leveraging the cell ontology to classify unseen cell
types. Nat Commun 2021, 12(1):5556.
145. Bernstein MN, Ma Z, Gleicher M, Dewey CN. CellO: comprehensive and
hierarchical cell type classification of human cells with the Cell Ontology.
iScience 2021, 24(1):101913.
146. Abdelaal T, Michielsen L, Cats D, Hoogduin D, Mei H, Reinders MJT,
Mahfouz A. A comparison of automatic cell identification methods for
single-​cell RNA sequencing data. Genome Biol 2019, 20(1):194.
147. Huang Q, Liu Y, Du Y, Garmire LX. Evaluation of cell type Annotation R
Packages on Single-​cell RNA-​seq Data. Genomics Proteomics Bioinformatics
2021, 19(2):267–​281.
148. Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, Chak S, Naikawadi RP,
Wolters PJ, Abate AR et al. Reference-​based analysis of lung single-​cell
sequencing reveals a transitional profibrotic macrophage. Nat Immunol
2019, 20(2):163–​172.
198 Next-Generation Sequencing Data Analysis

149. de Kanter JK, Lijnzaad P, Candelli T, Margaritis T, Holstege FCP.


CHETAH: a selective, hierarchical cell type identification method for single-​
cell RNA sequencing. Nucleic Acids Res 2019, 47(16):e95.
150. Cao ZJ, Wei L, Lu S, Yang DC, Gao G. Searching large-​scale scRNA-​seq
databases via unbiased cell embedding with Cell BLAST. Nat Commun 2020,
11(1):3458.
151. Hu J, Li X, Hu G, Lyu Y, Susztak K, Li M. Iterative transfer learning with
neural network for clustering and cell type classification in single-​cell RNA-​
seq analysis. Nat Mach Intell 2020, 2(10):607–​618.
152. Lieberman Y, Rokach L, Shay T. CaSTLe –​Classification of single cells by
transfer learning: Harnessing the power of publicly available single cell
RNA sequencing experiments to annotate new experiments. PLoS One 2018,
13(10):e0205499.
153. Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, Burgin G,
Delorey TM, Howitt MR, Katz Y et al. A single-​cell survey of the small intes-
tinal epithelium. Nature 2017, 551(7680):333–​339.
154. Hashimoto K, Kouno T, Ikawa T, Hayatsu N, Miyajima Y, Yabukami H,
Terooatea T, Sasaki T, Suzuki T, Valentine M et al. Single-​cell transcriptomics
reveals expansion of cytotoxic CD4 T cells in supercentenarians. Proc Natl
Acad Sci U S A 2019, 116(48):24242–​24251.
155. Cao Y, Lin Y, Ormerod JT, Yang P, Yang JYH, Lo KK. scDC: single cell differ-
ential composition analysis. BMC Bioinformatics 2019, 20(Suppl 19):721.
156. Jackson DA. Compositional data in community ecology: the paradigm or
peril of proportions? Ecology 1997, 78(3):929–​940.
157. Büttner M, Ostner J, Müller C, Theis F, Schubert B. scCODA is a Bayesian
model for compositional single-​ cell data analysis. Nat Commun 2021,
12(1):6876.
158. Kharchenko PV, Silberstein L, Scadden DT. Bayesian approach to single-​cell
differential expression analysis. Nat Methods 2014, 11(7):740–​742.
159. Finak G, McDavid A, Yajima M, Deng J, Gersuk V, Shalek AK, Slichter CK,
Miller HW, McElrath MJ, Prlic M et al. MAST: a flexible statistical frame-
work for assessing transcriptional changes and characterizing heterogeneity
in single-​cell RNA sequencing data. Genome Biol 2015, 16:278.
160. Delmans M, Hemberg M. Discrete distributional differential expression
(D3E) – a tool for gene expression analysis of single-​cell RNA-​seq data. BMC
Bioinformatics 2016, 17:110.
161. Korthauer KD, Chu LF, Newton MA, Li Y, Thomson J, Stewart R, Kendziorski
C. A statistical approach for identifying differential distributions in single-​
cell RNA-​seq experiments. Genome Biol 2016, 17(1):222.
162. Vu TN, Wills QF, Kalari KR, Niu N, Wang L, Rantalainen M, Pawitan Y. Beta-​
Poisson model for single-​cell RNA-​seq data analyses. Bioinformatics 2016,
32(14):2128–​2135.
163. Chen W, Li Y, Easton J, Finkelstein D, Wu G, Chen X. UMI-​count modeling
and differential expression analysis for single-​cell RNA sequencing. Genome
Biol 2018, 19(1):70.
164. Miao Z, Deng K, Wang X, Zhang X. DEsingle for detecting three types of
differential expression in single-​ cell RNA-​ seq data. Bioinformatics 2018,
34(18):3223–​3224.
Transcriptomics by Single-Cell RNA-Seq 199

165. Ye C, Speed TP, Salim A. DECENT: differential expression with capture


efficiency adjustmeNT for single-​ cell RNA-​ seq data. Bioinformatics 2019,
35(24):5155–​5162.
166. Das S, Rai SN. SwarnSeq: An improved statistical approach for differ-
ential expression analysis of single-​ cell RNA-​ seq data. Genomics 2021,
113(3):1308–​1324.
167. Van den Berge K, Roux de Bezieux H, Street K, Saelens W, Cannoodt R, Saeys
Y, Dudoit S, Clement L. Trajectory-​based differential expression analysis for
single-​cell sequencing data. Nat Commun 2020, 11(1):1201.
168. Wang T, Li B, Nelson CE, Nabavi S. Comparative analysis of differential
gene expression analysis tools for single-​cell RNA sequencing data. BMC
Bioinformatics 2019, 20(1):40.
169. Soneson C, Robinson MD. Bias, robustness and scalability in single-​cell dif-
ferential expression analysis. Nat Methods 2018, 15(4):255–​261.
170. Dal Molin A, Baruzzo G, Di Camillo B. Single-​ cell RNA-​ sequencing:
assessment of differential expression analysis methods. Front Genet 2017,
8:62.
171. Jaakkola MK, Seyednasrollah F, Mehmood A, Elo LL. Comparison of methods
to detect differentially expressed genes between single-​cell populations. Brief
Bioinform 2017, 18(5):735–​743.
172. Van den Berge K, Perraudeau F, Soneson C, Love MI, Risso D, Vert JP,
Robinson MD, Dudoit S, Clement L. Observation weights unlock bulk RNA-​
seq tools for zero inflation and single-​cell applications. Genome Biol 2018,
19(1):24.
173. Love MI, Huber W, Anders S. Moderated estimation of fold change and dis-
persion for RNA-​seq data with DESeq2. Genome Biol 2014, 15(12):550.
174. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for
differential expression analysis of digital gene expression data. Bioinformatics
2010, 26(1):139–​140.
175. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma
powers differential expression analyses for RNA-​sequencing and micro-
array studies. Nucleic Acids Res 2015, 43(7):e47.
176. Seyednasrollah F, Rantanen K, Jaakkola P, Elo LL. ROTS: reproducible RNA-​
seq biomarker detector-​prognostic markers for clear cell renal cell cancer.
Nucleic Acids Res 2016, 44(1):e1.
177. Vieth B, Parekh S, Ziegenhain C, Enard W, Hellmann I. A systematic evalu-
ation of single cell RNA-​seq analysis pipelines. Nat Commun 2019, 10(1):4667.
178. Saelens W, Cannoodt R, Todorov H, Saeys Y. A comparison of single-​cell tra-
jectory inference methods. Nat Biotechnol 2019, 37(5):547–​554.
179. Street K, Risso D, Fletcher RB, Das D, Ngai J, Yosef N, Purdom E, Dudoit
S. Slingshot: cell lineage and pseudotime inference for single-​ cell
transcriptomics. BMC Genomics 2018, 19(1):477.
180. Ji Z, Ji H. TSCAN: Pseudo-​time reconstruction and evaluation in single-​cell
RNA-​seq analysis. Nucleic Acids Res 2016, 44(13):e117.
181. Shin J, Berg DA, Zhu Y, Shin JY, Song J, Bonaguidi MA, Enikolopov G,
Nauen DW, Christian KM, Ming GL et al. Single-​cell RNA-​seq with water-
fall reveals molecular cascades underlying adult neurogenesis. Cell Stem Cell
2015, 17(3):360–​372.
200 Next-Generation Sequencing Data Analysis

182. Haghverdi L, Buttner M, Wolf FA, Buettner F, Theis FJ. Diffusion pseudotime
robustly reconstructs lineage branching. Nat Methods 2016, 13(10):845–​848.
183. Rashid S, Kotton DN, Bar-​Joseph Z. TASIC: determining branching models
from time series single cell data. Bioinformatics 2017, 33(16):2504–​2512.
184. Schiebinger G, Shu J, Tabaka M, Cleary B, Subramanian V, Solomon A, Gould
J, Liu S, Lin S, Berube P et al. Optimal-​Transport Analysis of Single-​Cell Gene
Expression identifies developmental trajectories in reprogramming. Cell
2019, 176(4):928–​943 e922.
185. Lin C, Bar-​ Joseph Z. Continuous-​ state HMMs for modeling time-​ series
single-​cell RNA-​Seq data. Bioinformatics 2019, 35(22):4707–​4715.
186. Tran TN, Bader GD. Tempora: cell trajectory inference using time-​series
single-​cell RNA sequencing data. PLoS Comput Biol 2020, 16(9):e1008205.
187. Lonnberg T, Svensson V, James KR, Fernandez-​Ruiz D, Sebina I, Montandon
R, Soon MS, Fogg LG, Nair AS, Liligeto U et al. Single-​cell RNA-​seq and com-
putational analysis using temporal mixture modelling resolves Th1/​Tfh fate
bifurcation in malaria. Sci Immunol 2017, 2(9):eaal2192.
188. LineagePulse (https://​git​hub.com/​Yosef​Lab/​Linea​gePu​lse)
189. La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V,
Lidschreiber K, Kastriti ME, Lonnerberg P, Furlan A et al. RNA velocity of
single cells. Nature 2018, 560(7719):494–​498.
190. Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity
to transient cell states through dynamical modeling. Nat Biotechnol 2020,
38(12):1408–​1414.
191. Herman JS, Sagar, Grun D. FateID infers cell fate bias in multipotent
progenitors from single-​ cell RNA-​ seq data. Nat Methods 2018,
15(5):379–​386.
192. Bendall SC, Davis KL, Amir el AD, Tadmor MD, Simonds EF, Chen TJ,
Shenfeld DK, Nolan GP, Pe’er D. Single-​cell trajectory detection uncovers
progression and regulatory coordination in human B cell development.
Cell 2014, 157(3):714–​725.
193. Setty M, Tadmor MD, Reich-​Zeliger S, Angel O, Salame TM, Kathail P,
Choi K, Bendall S, Friedman N, Pe’er D. Wishbone identifies bifurcating
developmental trajectories from single-​ cell data. Nat Biotechnol 2016,
34(6):637–​645.
194. Kim S, Scheffler K, Halpern AL, Bekritsky MA, Noh E, Kallberg M, Chen
X, Kim Y, Beyter D, Krusche P et al. Strelka2: fast and accurate calling of
germline and somatic variants. Nat Methods 2018, 15(8):591–​594.
195. Rodriguez-​Meira A, Buck G, Clark SA, Povinelli BJ, Alcolea V, Louka E,
McGowan S, Hamblin A, Sousos N, Barkas N et al. Unravelling Intratumoral
Heterogeneity through High-​ Sensitivity Single-​
Cell Mutational Analysis
and Parallel RNA Sequencing. Mol Cell 2019, 73(6):1292–​1305 e1298.
196. Pysam (https://​git​hub.com/​pysam-​dev​elop​ers/​pysam)
197. Fasterius E, Uhlen M, Al-​Khalili Szigyarto C. Single-​cell RNA-​seq variant
analysis for exploration of genetic heterogeneity in cancer. Sci Rep 2019,
9(1):9524.
198. Zafar H, Wang Y, Nakhleh L, Navin N, Chen K. Monovar: single-​nucleotide
variant detection in single cells. Nat Methods 2016, 13(6):505–​507.
199. Schnepp PM, Chen M, Keller ET, Zhou X. SNV identification from single-​cell
RNA sequencing data. Hum Mol Genet 2019, 28(21):3569–​3583.
Transcriptomics by Single-Cell RNA-Seq 201

200. Poirion O, Zhu X, Ching T, Garmire LX. Using single nucleotide variations
in single-​cell RNA-​seq to identify subpopulations and genotype-​phenotype
linkage. Nat Commun 2018, 9(1):4892.
201. Fangal VD. CTAT Mutations: A Machine Learning Based RNA-​Seq Variant
Calling Pipeline Incorporating Variant Annotation, Prioritization, and
Visualization. 2020.
202. Huang X, Huang Y. Cellsnp-​lite: an efficient tool for genotyping single cells.
Bioinformatics 2021, 37(23):4569–​4571.
203. Liu F, Zhang Y, Zhang L, Li Z, Fang Q, Gao R, Zhang Z. Systematic compara-
tive analysis of single-​nucleotide variant detection methods from single-​cell
RNA sequencing data. Genome Biol 2019, 20(1):242.
204. Chung W, Eum HH, Lee HO, Lee KM, Lee HB, Kim KT, Ryu HS, Kim S, Lee
JE, Park YH et al. Single-​cell RNA-​seq enables comprehensive tumour and
immune cell profiling in primary breast cancer. Nat Commun 2017, 8:15081.
205. Fan J, Lee HO, Lee S, Ryu DE, Lee S, Xue C, Kim SJ, Kim K, Barkas N, Park PJ
et al. Linking transcriptional and genetic tumor heterogeneity through allele
analysis of single-​cell RNA-​seq data. Genome Res 2018, 28(8):1217–​1227.
206. Serin Harmanci A, Harmanci AO, Zhou X. CaSpER identifies and visualizes
CNV events by integrative analysis of single-​cell or bulk RNA-​sequencing
data. Nat Commun 2020, 11(1):89.
207. inferCNV of the Trinity CTAT Project (https://​git​hub.com/​bro​adin​stit​ute/​
infer​CNV)
208. Muller S, Cho A, Liu SJ, Lim DA, Diaz A. CONICS integrates scRNA-​seq with
DNA sequencing to map gene expression to tumor sub-​clones. Bioinformatics
2018, 34(18):3217–​3219.
209. van de Geijn B, McVicker G, Gilad Y, Pritchard JK. WASP: allele-​specific soft-
ware for robust molecular quantitative trait locus discovery. Nat Methods
2015, 12(11):1061–​1063.
210. Borel C, Ferreira PG, Santoni F, Delaneau O, Fort A, Popadin KY, Garieri M,
Falconnet E, Ribaux P, Guipponi M et al. Biased allelic expression in human
primary fibroblast single cells. Am J Hum Genet 2015, 96(1):70–​80.
211. Song Y, Botvinnik OB, Lovci MT, Kakaradov B, Liu P, Xu JL, Yeo GW. Single-​
cell alternative splicing analysis with expedition reveals splicing dynamics
during neuron differentiation. Mol Cell 2017, 67(1):148–​161 e145.
212. Huang Y, Sanguinetti G. BRIE: transcriptome-​wide splicing quantification in
single cells. Genome Biol 2017, 18(1):123.
213. Huang Y, Sanguinetti G. BRIE2: computational identification of splicing
phenotypes from single-​cell transcriptomic experiments. Genome Biol 2021,
22(1):251.
214. Matsumoto H, Hayashi T, Ozaki H, Tsuyuzaki K, Umeda M, Iida T,
Nakamura M, Okano H, Nikaido I. An NMF-​based approach to discover
overlooked differentially expressed gene regions from single-​cell RNA-​seq
data. NAR Genom Bioinform 2019, 2(1):lqz020.
215. Ling JP, Wilks C, Charles R, Leavey PJ, Ghosh D, Jiang L, Santiago CP, Pang
B, Venkataraman A, Clark BS et al. ASCOT identifies key regulators of neur-
onal subtype-​specific splicing. Nat Commun 2020, 11(1):137.
216. Ozaki H, Hayashi T, Umeda M, Nikaido I. Millefy: visualizing cell-​to-​cell
heterogeneity in read coverage of single-​cell RNA sequencing datasets. BMC
Genomics 2020, 21(1):177.
202 Next-Generation Sequencing Data Analysis

217. Wen WX, Mead AJ, Thongjuea S. VALERIE: Visual-​ based inspection of
alternative splicing events at single-​cell resolution. PLoS Comput Biol 2020,
16(9):e1008195.
218. Hu Y, Wang K, Li M. Detecting differential alternative splicing events in
scRNA-​seq with or without unique molecular identifiers. PLoS Comput Biol
2020, 16(6):e1007925.
219. Benegas G, Fischer J, Song YS. Robust and annotation-​free analysis of alter-
native splicing across diverse cell types in mice. Elife 2022, 11:e73520.
220. Liu S, Zhou B, Wu L, Sun Y, Chen J, Liu S. Single-​cell differential splicing
analysis reveals high heterogeneity of liver tumor-​infiltrating T cells. Sci Rep
2021, 11(1):5325.
221. Aibar S, Gonzalez-​ Blas CB, Moerman T, Huynh-​ Thu VA, Imrichova
H, Hulselmans G, Rambow F, Marine JC, Geurts P, Aerts J et al.
SCENIC: single-​cell regulatory network inference and clustering. Nat Methods
2017, 14(11):1083–​1086.
222. Matsumoto H, Kiryu H, Furusawa C, Ko MSH, Ko SBH, Gouda N, Hayashi
T, Nikaido I. SCODE: an efficient regulatory network inference algorithm
from single-​ cell RNA-​ Seq during differentiation. Bioinformatics 2017,
33(15):2314–​2321.
223. Matsumoto H, Kiryu H. SCOUP: a probabilistic model based on the Ornstein-​
Uhlenbeck process to analyze single-​cell expression data during differenti-
ation. BMC Bioinformatics 2016, 17(1):232.
224. Chan TE, Stumpf MPH, Babtie AC. Gene Regulatory Network Inference
from Single-​Cell Data Using Multivariate Information Measures. Cell Syst
2017, 5(3):251–​267 e253.
225. Specht AT, Li J. LEAP: constructing gene co-​expression networks for single-​
cell RNA-​sequencing data using pseudotime ordering. Bioinformatics 2017,
33(5):764–​766.
226. Liu H, Li P, Zhu M, Wang X, Lu J, Yu T. Nonlinear Network Reconstruction
from Gene Expression Data Using Marginal Dependencies Measured by
DCOL. PLoS One 2016, 11(7):e0158247.
227. Cordero P, Stuart JM. Tracing Co-​Regulatory Network Dynamics in Noisy,
Single-​Cell Transcriptome Trajectories. Pac Symp Biocomput 2017, 22:576–​587.
228. Aubin-​Frankowski PC, Vert JP. Gene regulation inference from single-​cell
RNA-​ seq data with linear differential equations and velocity inference.
Bioinformatics 2020, 36(18):4774–​4780.
229. Huynh-​ Thu VA, Irrthum A, Wehenkel L, Geurts P. Inferring regulatory
networks from expression data using tree-​based methods. PLoS One 2010,
5(9):e12776.
230. Moerman T, Aibar Santos S, Bravo Gonzalez-​Blas C, Simm J, Moreau Y, Aerts
J, Aerts S. GRNBoost2 and Arboreto: efficient and scalable inference of gene
regulatory networks. Bioinformatics 2019, 35(12):2159–​2161.
231. Woodhouse S, Piterman N, Wintersteiger CM, Gottgens B, Fisher J. SCNS: a
graphical tool for reconstructing executable regulatory networks from single-​
cell genomic data. BMC Syst Biol 2018, 12(1):59.
232. Lim CY, Wang H, Woodhouse S, Piterman N, Wernisch L, Fisher J, Gottgens
B. BTR: training asynchronous Boolean models using single-​cell expression
data. BMC Bioinformatics 2016, 17(1):355.
Transcriptomics by Single-Cell RNA-Seq 203

233. Pratapa A, Jalihal AP, Law JN, Bharadwaj A, Murali TM. Benchmarking
algorithms for gene regulatory network inference from single-​ cell
transcriptomic data. Nat Methods 2020, 17(2):147–​154.
234. Chen S, Mar JC. Evaluating methods of inferring gene regulatory networks
highlights their lack of performance for single cell gene expression data.
BMC Bioinformatics 2018, 19(1):232.
235. Nguyen H, Tran D, Tran B, Pehlivan B, Nguyen T. A comprehensive survey
of regulatory network inference methods using single-​cell RNA sequencing
data. Brief Bioinform 2020, 22(3):bbaa190.
236. Kang Y, Thieffry D, Cantini L. Evaluating the Reproducibility of Single-​Cell
Gene Regulatory Network Inference Algorithms. Front Genet 2021, 12:617282.
237. Dai H, Li L, Zeng T, Chen L. Cell-​specific network constructed by single-​cell
RNA sequencing data. Nucleic Acids Res 2019, 47(11):e62.
238. Li L, Dai H, Fang Z, Chen L. c-​CSN: Single-​cell RNA Sequencing Data
Analysis by Conditional Cell-​ specific Network. Genomics Proteomics
Bioinformatics 2021, 19(2):319–​329.
239. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation
network analysis. BMC Bioinformatics 2008, 9:559.
9 
Small RNA Sequencing

Small RNAs play an important role in regulating gene expression in both


the cytoplasm and the nucleus through inducing both post-​transcriptional
and transcriptional gene silencing mechanisms. In addition to RNAi, some
studies also show that some small RNAs can increase gene expression via a
mechanism called RNA activation (or RNAa) [1]. Through these regulatory
activities, small RNAs are involved in many cellular processes, affect growth
and development, and if their own expression goes awry, lead to diseases
such as cancer and Alzheimer’s disease.
As introduced in Chapter 3, the major categories of small RNAs in
cells include miRNAs, siRNAs, and piRNAs. Among these three types
of small RNAs, miRNAs are so far the most studied. A total of 38,589
miRNA loci have been catalogued in 271 species at the time of this writing
in miRBase (release 22.1), the gold-​standard database for miRNAs. It
has been estimated that a typical mammalian cell contains hundreds of
miRNA species, each of which regulates transcripts from multiple genes.
The expression of these miRNAs is cell-​and tissue-​specific, and dynam-
ically regulated based on cellular state. Mutations or methylations in
miRNA genes often lead to dysregulation in their expression. Studying
the expression of miRNAs and other small RNAs is an important aspect
of studying their roles in biological processes and diseases. Compared to
other small RNA expression analysis methods, such as microarray and
qPCR, NGS has a broader dynamic range for measuring small RNAs
even at extremely high or low levels, single-​base resolution to differen-
tiate closely related small RNA molecules, the ability to study organisms
without a currently available genome assembly, and the capability to dis-
cover novel small RNA species.
On new small RNA discovery, although from human and other model
organisms the community has catalogued thousands of miRNAs and other
small RNA species, more remain to be found. For less studied species, the
number of known small RNAs is still low. Many in silico miRNA prediction
algorithms have been developed, but their predictions have to be validated
with experimental evidence. Small-​RNA sequencing, through interrogating

DOI: 10.1201/9780429329180-12 205


206 Next-Generation Sequencing Data Analysis

the entire pool of small RNAs, provides an excellent tool for novel miRNA
discovery and experimental validation of computational predictions.
Furthermore, small RNA sequencing offers an assumption-​free, comprehen-
sive analysis of the small RNA transcriptome in biological targets, including
differential expression between conditions. In general, small RNA sequen-
cing data analysis shares much commonality with the analysis of RNA-​seq
data (Chapter 7). In the meantime, some aspects of small RNA sequencing
data analysis are unique and mostly focused on in this chapter.

9.1 Small RNA NGS Data Generation and Upstream Processing


9.1.1 Data Generation
Since sequencing analysis of small RNAs in the transcriptome is similar to
mRNA analysis, the experimental aspects detailed in Chapter 7 on factorial
design, replication and randomization, and sample collection equally apply
here and are therefore not repeated. Mature miRNA species, generated as a
result of Dicer and Argonaute processing (Figure 9.1), have an average size
of 22 nucleotides. Small RNA molecules can be purified from cells or tissues,
while total RNA extracts that retain small RNA species work equally well and
are often used. A size selection step in the sequencing library construction
process removes larger RNA molecules in total RNA extracts. Furthermore,
the small RNA sequencing library construction process takes advantage of
the particular end structure on small RNAs, which are absent on mRNAs.

Sequence
Reads

miRNA-miRNA*
Pre-miRNA Loop
Duplex

Dicer *Argonaut *
Cleavage Processing
and Deep
Sequencing

FIGURE 9.1
Deep sequencing of mature miRNAs after Dicer and Argonaute processing. Dicer cleaves a short
stem-​loop structure out of pre-​miRNA to form the miRNA:miRNA* duplex. Upon loading into
RISC, Argonaute unwinds the duplex and uses one strand as guide for gene silencing while
discards the other strand (the star strand). While the short stem-​loop and star strand sequences
are usually degraded, they may still generate sequencing signals, because of undegraded
residues or the fact that they may exist to perform other functions (e.g., the star strand is
sometimes functional).
Small RNA Sequencing 207

Canonical mature small RNAs have a monophosphate group at the 5’ end


and a hydroxyl group at the 3’ end, which is derived from the action of small
RNA processing enzymes such as Dicer.
Many small RNA sequencing library construction processes start
with ligation of adapter sequences to their 3’ and 5’ ends. The universal
adapter sequences provide anchoring for subsequent reverse transcription,
and then PCR amplification. Among these steps, the initial ligation step
has been found to introduce most bias to the process [2]. To counter the
biases introduced during ligation as well as subsequent PCR amplifica-
tion, alternative strategies have been devised and employed. The strategies
for mitigation of the ligation bias include use of modified adapters that
carry randomized nucleotides at the ligation boundary [3], and ligation-​
free procedures such as the SMART polyadenylation and template switch
technology followed by PCR amplification, or probe capture-​based target
miRNA sequencing. To mitigate PCR bias, unique molecular identifiers
(UMIs) can be used. To select an appropriate library construction procedure,
the specific needs of the project need to be considered, as all currently avail-
able protocols have strengths and weaknesses based on currently available
benchmark studies [2, 4].
Because of their short length, constructed small RNA sequencing libraries
do not need to be sequenced very long. The actual read length depends on
the configuration of library constructs and whether the index sequences are
read in the same pass or as a separate reading step. In the current version of
the Illumina small RNA sequencing protocol that reads index sequences in
a second pass, 50 cycles of sequencing can be enough. Sequencing depth is
another key factor in the data generation process that determines the power
of differential expression analysis and novel small RNA discovery. While
this depends on sample source as small RNA amount and composition
vary greatly with cell type and species, in general 2–​3 million aligned reads
(or 4–​5 million raw unmapped reads) should offer enough confidence for
most studies. A study has shown that coverage higher than 5 million reads
contributes little to the detection of new small RNA species [5].

9.1.2 Preprocessing
After obtaining sequencing reads and demultiplexing, the reads generated
from each sample need to be first checked for quality using the QC tools
introduced in Chapter 5 such as FastQC, NGS QC Toolkit, and fastp, or spe-
cifically developed miRNA-​seq data QC tools including miRTrace [6] and
mirnaQC [7]. Besides the typical NGS data QC metrics, specific miRNA-​seq
QC tools often provide additional features. For example, mirnaQC provides
quality measures on miRNA yield and the fraction of putative degradation
products (e.g., rRNA fragments) in both absolute values and relative ranks
in comparison to a reference collection of 36,000 published datasets. Because
208 Next-Generation Sequencing Data Analysis

small RNA libraries are usually sequenced longer than the actual lengths of
the small RNA inserts, the 3’ adapter sequence is often part of the generated
sequence reads and therefore should also be trimmed off. The trimming can
be carried out with stand-alone tools such as Cutadapt and Trimmomatic,
or utilities in the NGS QC Toolkit or fastp. Adapter trimming can also be
conducted coincidentally with mapping, as some mappers provide such an
option, or using data preprocessing modules within some small RNA data
analysis tools (to be covered next).

9.1.3 Mapping
For mapping small RNA sequencing reads to a reference genome, short read
aligners introduced in Chapter 5, such as Bowtie/​Bowtie2, BWA, Novoalign,
or SOAP/​SOAP2, or RNA-​seq aligner in Chapter 7, such as STAR, can be
used. Among these aligners, Novoalign offers the option of stripping off
adapter sequences in the mapping command. As for the reference genome,
the most recent assembly should always be used. Because of the short target
read length, the number of allowed mismatches should be set as 1. To speed
up the mapping process, a multi-​threading parameter, which enables the use
of multiple CPU cores, can be used if the aligner supports it. After mapping,
reads that are aligned to unique regions are then searched against small RNA
databases to establish their identities (see next section), while those that
are mapped to a large number (e.g., >5,000) of genomic locations should be
removed from further analysis.
Besides the aforementioned general tools for small RNA reads
preprocessing and mapping, tools have also been developed specially for
small RNA-​seq analysis, such as miRDeep/​miRDeep2 [8], sRNAtoolbox [9],
ShortStack [10], sRNAnalyzer [11], miRge [12], and miRMaster [13]. Among
these tools, sRNAtoolbox is a collection of small RNA-​seq data tools for dif-
ferential expression analysis and other downstream analyses. Its center piece
is sRNAbench, which replaces the previously widely used miRanalyzer. It
provides functions such as data preprocessing, genome mapping using
Bowtie, visualization of genome mapped reads, expression profiling, etc.
While the mapping of small RNA reads to a reference genome is similar
to the mapping in RNA-​seq as covered in Chapter 7, some characteristics of
small RNAs, mostly their short length and post-​transcriptional editing (see
next), present different challenges to the small RNA read mapping process.
Because of their short length, sizeable numbers of small RNA reads are usu-
ally mapped to more than one genomic region. In comparison, this issue is
minimal for RNA-​seq data, as longer and sometimes paired-​end reads greatly
increase specificity. The easiest way to deal with multi-​mapped small RNA
reads is to simply ignore them, but this leads to the loss of great amounts of
data. A more commonly used approach is to assign them to one of the mapped
positions randomly, while an alternative approach is to report them to all
possible positions. More sophisticated algorithms have also been developed
Small RNA Sequencing 209

in an effort to avoid the precision or sensitivity pitfalls of these approaches.


For example, ShortStack employs local weighting, which uses local genomic
context to guide placement of multi-​mapped miRNA-​seq reads.
Post-​transcriptional editing, on the other hand, leads to the generation of
isomiRs [14], i.e., isoforms of canonical miRNAs that resemble but never-
theless vary from the reference miRNA annotated in miRBase. The isomiRs
have various forms of variations from the canonical sequence, including
alternative 3’ (more often) and 5’ termini, and nucleotide substitutions in
the body sequence. Since their discovery, which itself is attributed to small
RNA sequencing, isomiRs have been shown to have functional importance
[15]. While the discovery of isomiRs is more recent, many newer tools cover
isomiRs, including miRge, sRNAnalyzer, and sRNAbench.

9.1.4 Identification of Known and Putative Small RNA Species


To identify currently known small RNA species, the mapped reads need to
be searched against the most recent version of miRBase or other small RNA
databases (such as piRNABank, piRBase, and piRNAclusterDB for piRNAs).
Reads with no matches in these databases can then be searched against
other databases (Rfam, repeat, and mRNA) to determine if they are degrad-
ation products of ncRNAs, genomic repeats, and mRNAs. The previously
mentioned small RNA-​seq data analysis tools all provide these database
search capabilities.
To discover potentially novel miRNA species, mapped reads that do not
match known miRNAs and sequences in the other databases are submitted
to algorithms such as sRNAbench and miRDeep2, which are designed to
search for putative miRNAs. The approach used by miRDeep2 takes into
consideration the biogenic process of miRNAs. It first identifies poten-
tial miRNA precursor coding regions out of the genomic regions that are
clustered with the mapped reads. RNA secondary structures are then
predicted on these identified regions using RNA folding software, and
examined to see if they resemble a typical miRNA hairpin structure seen in
pri-​miRNA molecules and if they are thermodynamically stable. Putative
miRNA species are called if the reads fall into stable hairpins in an expected
manner, along with other evidences such as reads from the star strand.
Similarly, sRNAbench also uses both structural and biogenic features to
predict novel miRNAs [16].

9.1.5 Normalization
Before identifying differentially expressed small RNAs, read counts for each
small RNA species in the samples need to be normalized. The goal of normal-
ization is to make the samples directly comparable by removing unwanted
sample-​specific variations, which are usually due to differences in library size
and therefore sequencing depth. The normalization approaches used in bulk
210 Next-Generation Sequencing Data Analysis

RNA-​seq as detailed in Chapter 7 can be similarly applied here. The general


assumption for most of the normalization approaches, that the majority of
small RNAs stay constant between conditions, seems to hold. For the total
read-​count-​based normalization, since all small RNAs are similarly short in
size, the RPKM normalization can be simplified as RPM (reads per million).
This method, however, has been found to be inadequate in some bench-
mark studies [17–​19]. Other more sophisticated normalization approaches
introduced in Chapter 7, including RLE used by DESeq/​DESeq2 and TMM
used by edgeR, have been found to work well for identification of differen-
tially expressed small RNAs.

9.2 Identification of Differentially Expressed Small RNAs


The packages and tests introduced for RNA-​ seq differential expression
analysis in Chapter 7 can also be directly used for small RNA analysis. For
example, DESeq2, edgeR, limma, DEGseq, and NOISeq have been widely
employed on the identification of differentially expressed small RNAs.
In terms of implementation, these methods can be either paired with or
integrated into the tools specifically designed for small RNA-​seq data ana-
lysis as detailed in Section 9.1.3. For example, miRDeep2 can be used first
to identify and quantify known and novel miRNAs, and the raw counts
of identified miRNAs can then be input into edgeR or limma for DE ana-
lysis. To compute differential expression, miRge integrates with DESeq2.
The sRNAtoolbox has a module called sRNAde specially developed for DE
analysis. Using sRNAbench-​generated or user-​provided expression matrix
as input, this module detects differentially expressed small RNAs with the
use of five methods: edgeR, DESeq, DESeq2, NOISeq, and Student’s t-​test.
Besides DE results generated from the individual methods, sRNAde also
provides consensus DE results from the five methods. FDR is provided by all
of these methods for multiple testing correction.

9.3 Functional Analysis of Identified Known Small RNAs


To perform functional analysis of differentially expressed small RNAs, their
gene targets need to be predicted first. A number of tools are available for
this task, including TargetScan [20], miRanda [21], mirSVR [22], miRDB
[23], miRWalk [24], PicTar [25], PITA [26], RNA22 [27], RNAhybrid [28],
DeepMirTar [29], and the DNA intelligent analysis (DIANA) applications
microT-​CDS and microT [30]. These tools predict target genes based on two
general approaches, with one based on characteristics of miRNA-​mRNA
Small RNA Sequencing 211

interaction, and the other on inference from machine learning models.


The first approach makes predictions on the basis of seed pairing, thermo-
dynamic stability, sequence conservation, and 3’-​UTR structural accessibility.
Many of the methods above use this approach. For example, miRanda, one of
the earliest developed and widely used methods, makes predictions based on
miRNA-​mRNA complementarity pattern, location of the binding site in the
mRNA, binding energy, and miRNA evolutionary conservation. The second
machine-​learning-​based approach applies artificial intelligence to learn from
known miRNA-​mRNA duplexes to make new predictions. This approach
uses features extracted from validated miRNA-​ mRNA interactions, plus
negative dataset, to train a classifier model that can make distinctions and
predict new miRNA targets. This approach has the ability to automatically
improve based on new results. Examples of methods using this approach
include DIANA-​microT-​CDS, mirSVR, and DeepMirTar.
Despite the progress made on miRNA target gene prediction, miRNA
target gene prediction is still no easy task because of the small size of the
miRNA-​ mRNA binding area, often imperfect complementarity of the
binding, and sometimes lack of conservation [31]. Once a list of potential
target genes is generated, functional analysis, such as Gene Ontology (GO)
and pathway analysis, can be conducted using the approaches detailed in
Chapter 7. In addition, for pathway analysis, a list of miRNAs can also be
directly uploaded to the DIANA miRPath web server to a generate a list of
biological pathways that are significantly enriched with the miRNAs’ target
genes, which are predicted with DIANA-​microT-​CDS or documented with
existing experimental evidence [32].

References
1. Huang V, Qin Y, Wang J, Wang X, Place RF, Lin G, Lue TF, Li LC. RNAa is
conserved in mammalian cells. PLoS One 2010, 5(1):e8848.
2. Androvic P, Benesova S, Rohlova E, Kubista M, Valihrach L. Small RNA-​
sequencing for analysis of circulating mirnas: benchmark study. J Mol Diagn
2022, 24(4):386–​394.
3. Baran-​Gale J, Kurtz CL, Erdos MR, Sison C, Young A, Fannin EE, Chines PS,
Sethupathy P. Addressing bias in small RNA library preparation for sequen-
cing: a new protocol recovers microRNAs that evade capture by current
methods. Front Genet 2015, 6:352.
4. Benesova S, Kubista M, Valihrach L. Small RNA-​sequencing: approaches and
considerations for miRNA analysis. Diagnostics (Basel) 2021, 11(6):964.
5. Metpally RP, Nasser S, Malenica I, Courtright A, Carlson E, Ghaffari L,
Villa S, Tembe W, Van Keuren-​Jensen K. Comparison of analysis tools for
miRNA high throughput sequencing using nerve crush as a model. Front
Genet 2013, 4:20.
212 Next-Generation Sequencing Data Analysis

6. Kang W, Eldfjell Y, Fromm B, Estivill X, Biryukova I, Friedlander MR. miRTrace


reveals the organismal origins of microRNA sequencing data. Genome Biol
2018, 19(1):213.
7. Aparicio-​Puerta E, Gomez-​Martin C, Giannoukakos S, Medina JM, Marchal
JA, Hackenberg M. mirnaQC: a webserver for comparative quality control of
miRNA-​seq data. Nucleic Acids Res 2020, 48(W1):W262–​W267.
8. Friedlander MR, Mackowiak SD, Li N, Chen W, Rajewsky N. miRDeep2
accurately identifies known and hundreds of novel microRNA genes in seven
animal clades. Nucleic Acids Res 2012, 40(1):37–​52.
9. Aparicio-​ Puerta E, Gomez-​ Martin C, Giannoukakos S, Medina JM,
Scheepbouwer C, Garcia-​Moreno A, Carmona-​Saez P, Fromm B, Pegtel M,
Keller A et al. sRNAbench and sRNAtoolbox 2022 update: accurate miRNA
and sncRNA profiling for model and non-​model organisms. Nucleic Acids Res
2022, 50(W1):W710–​W717.
10. Johnson NR, Yeoh JM, Coruh C, Axtell MJ. Improved placement of multi-​
mapping small RNAs. G3 2016, 6(7):2103–​2111.
11. Wu X, Kim TK, Baxter D, Scherler K, Gordon A, Fong O, Etheridge A, Galas
DJ, Wang K. sRNAnalyzer-​a flexible and customizable small RNA sequencing
data analysis pipeline. Nucleic Acids Res 2017, 45(21):12140–​12151.
12. Patil AH, Halushka MK. miRge3.0: a comprehensive microRNA and tRF
sequencing analysis pipeline. NAR Genom Bioinform 2021, 3(3):lqab068.
13. Fehlmann T, Kern F, Laham O, Backes C, Solomon J, Hirsch P, Volz C, Muller
R, Keller A. miRMaster 2.0: multi-​species non-​coding RNA sequencing ana-
lyses at scale. Nucleic Acids Res 2021, 49(W1):W397–​W408.
14. Morin RD, O’Connor MD, Griffith M, Kuchenbauer F, Delaney A, Prabhu AL,
Zhao Y, McDonald H, Zeng T, Hirst M et al. Application of massively parallel
sequencing to microRNA profiling and discovery in human embryonic stem
cells. Genome Res 2008, 18(4):610–​621.
15. Tomasello L, Distefano R, Nigita G, Croce CM. The MicroRNA Family Gets
Wider: The IsomiRs Classification and Role. Front Cell Dev Biol 2021, 9:668648.
16. Barturen G, Rueda A, Hamberg M, Alganza A, Lebron R, Kotsyfakis M, Shi B-​
J, Koppers-​Lalic D, Hackenberg M. sRNAbench: profiling of small RNAs and
its sequence variants in single or multi-​species high-​throughput experiments.
Methods Next-​Generation Seq. 2014, 1:21–​31.
17. Garmire LX, Subramaniam S. Evaluation of normalization methods in mam-
malian microRNA-​Seq data. RNA 2012, 18(6):1279–​1288.
18. Dillies MA, Rau A, Aubert J, Hennequet-​Antier C, Jeanmougin M, Servant
N, Keime C, Marot G, Castel D, Estelle J et al. A comprehensive evaluation of
normalization methods for Illumina high-​throughput RNA sequencing data
analysis. Brief Bioinform 2013, 14(6):671–​683.
19. Tam S, Tsao MS, McPherson JD. Optimization of miRNA-​ seq data
preprocessing. Brief Bioinform 2015, 16(6):950–​963.
20. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target
sites in mammalian mRNAs. Elife 2015, 4:e05005.
21. Enright AJ, John B, Gaul U, Tuschl T, Sander C, Marks DS. MicroRNA targets
in Drosophila. Genome Biol 2003, 5(1):R1.
22. Betel D, Koppal A, Agius P, Sander C, Leslie C. Comprehensive modeling of
microRNA targets predicts functional non-​conserved and non-​canonical sites.
Genome Biol 2010, 11(8):R90.
Small RNA Sequencing 213

23. Chen Y, Wang X. miRDB: an online database for prediction of functional


microRNA targets. Nucleic Acids Res 2020, 48(D1):D127–​D131.
24. Sticht C, De La Torre C, Parveen A, Gretz N. miRWalk: An online resource for
prediction of microRNA binding sites. PLoS One 2018, 13(10):e0206239.
25. Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P,
da Piedade I, Gunsalus KC, Stoffel M et al. Combinatorial microRNA target
predictions. Nat Genet 2005, 37(5):495–​500.
26. Kertesz M, Iovino N, Unnerstall U, Gaul U, Segal E. The role of site accessi-
bility in microRNA target recognition. Nat Genet 2007, 39(10):1278–​1284.
27. Miranda KC, Huynh T, Tay Y, Ang YS, Tam WL, Thomson AM, Lim B, Rigoutsos
I. A pattern-​based method for the identification of microRNA binding sites
and their corresponding heteroduplexes. Cell 2006, 126(6):1203–​1217.
28. Rehmsmeier M, Steffen P, Hochsmann M, Giegerich R. Fast and effective pre-
diction of microRNA/​target duplexes. RNA 2004, 10(10):1507–​1517.
29. Wen M, Cong P, Zhang Z, Lu H, Li T. DeepMirTar: a deep-​learning approach
for predicting human miRNA targets. Bioinformatics 2018, 34(22):3781–​3787.
30. Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T,
Reczko M, Filippidis C, Dalamagas T, Hatzigeorgiou AG. DIANA-​microT web
server v5.0: service integration into miRNA functional analysis workflows.
Nucleic Acids Res 2013, 41(Web Server issue):W169–​173.
31. Ritchie W, Flamant S, Rasko JE. Predicting microRNA targets and
functions: traps for the unwary. Nat Methods 2009, 6(6):397–​398.
32. Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni
D, Vergoulis T, Dalamagas T, Hatzigeorgiou AG. DIANA-​ miRPath
v3.0: deciphering microRNA function with experimental support. Nucleic
Acids Res 2015, 43(W1):W460–​466.
10 
Genotyping and Variation Discovery
by Whole Genome/​Exome Sequencing

Detection of genomic variation among individuals of a population is among


the most frequent applications of NGS. Genome sequence heterogeneity is
prevalent in a naturally occurring population, which cannot be captured by
the current use of a single reference genome for a species. Genomic variant
cataloging projects in many countries, such as the All of Us Program in the
United States that aims to sequence one million genomes, underscore the
importance of genomic variation discovery. Locating genomic sequence
variations that correlate with disease predisposition or drug response, and
establishing genotypic basis of various phenotypes, have become common
focuses of many NGS studies in biomedical and life science research. Besides
variations carried through the germline for generations, NGS has also been
applied to identify de novo germline and somatic mutations, which occur
more frequently than previously expected and underlie numerous human
diseases including cancer and neurodegenerative diseases [1–​3].
Detecting from NGS data the various forms of genomic variations/​
mutations detailed in Chapter 2, including SNVs, indels, and SVs, is not an
easy task. The primary challenge is to differentiate true sequence variations/​
mutations from false positives caused by sequencing errors and artifacts
generated in basecalling and sequence alignment. It is, therefore, important
to generate high-​quality sequencing data before performing data analysis.
Equally importantly, sensitive and yet specific variant/​ mutant calling
algorithms are required to achieve high accuracy in genomic variation and
mutation discovery. This chapter first provides details on data preprocessing,
alignment, realignment, and recalibration, then focuses on methods for the
detection of germline and somatic SNVs/​indels and SVs, followed by anno-
tation of identified variants. Figure 10.1 shows an overview of the data ana-
lysis pipeline.

DOI: 10.1201/9780429329180-13 215


216 Next-Generation Sequencing Data Analysis

Data Preprocessing & QC

Read Mapping

Local Realignment

Base Quality Recalibration

Variant Calling

Germline SNV Somatic Structural Copy Number


& Short Indel Mutation Variant Variant

Variant Annotation

FIGURE 10.1
General workflow for genotyping and variation discovery from resequencing data.

10.1 Data Preprocessing, Mapping, Realignment,


and Recalibration
Besides the general data preprocessing and QC steps introduced in Chapter 5,
such as examining sequencing data quality, removing low-​quality and dupli-
cate reads, additional steps are needed for variant calling. The read mapping
step requires the use of highly sensitive alignment algorithm such as BWA-​
MEM, Bowtie2, and minimap2. After examining mapping quality, reads with
low-​quality mapping scores need to be filtered out. For paired-​end reads,
they should map to the reference genome as pairs at the expected interval and
those that do not show the expected pattern should be filtered out as well.
After the initial alignment, realignment around indels usually leads to
improvement in mapping results. This is usually due to the fact that short
indels, especially those at the ends of reads, often cause problems in the ini-
tial alignment process. To realign around the indel regions, the original BAM
Whole Genome/Exome Sequencing 217

file is first processed to identify where realignment is needed using tools


such as the GATK RealignerTargetCreator. In this process, using a known
set of indels (such as those in dbSNP) can speed up the process and improve
accuracy. After the target regions for realignment are identified, programs
such as the GATK IndelRealigner can be employed to conduct the realign-
ment. At the end of this process, a new BAM file is generated containing
realigned reads.
Prior to variant calling, the original base​call quality scores should also
be recalibrated to further improve data quality. This base quality score
recalibration can be conducted with tools such as the GATK BaseRecalibrator
and ApplyBQSR. These tools recalibrate raw quality values using covariate-​
aware base quality score recalibration algorithms, which adjust for covariates,
such as machine sequencing cycle and local sequence context, that are known
to affect sequencing signal and basecall quality. To carry out the recalibration,
a model of covariation is first analyzed and built by BaseRecalibrator when
using GATK, which is then used by ApplyBQSR to recalibrate the data.
Variant calling based on the recalibrated data has higher accuracy and cuts
down on the number of false positives.

10.2 Single Nucleotide Variant (SNV) and Short Indel Calling


10.2.1 Germline SNV and Indel Calling
In general, variant calls are affected by a number of factors (Figure 10.2).
These factors include: 1) basecall quality, 2) mapping quality, 3) single vs.
paired-​end sequencing, 4) read length, 5) depth of coverage, and 6) sequence
context. Because of errors or uncertainties that occur in the steps of sequen-
cing/​basecalling/​mapping, there are almost always certain levels of uncer-
tainty associated with each variant call. To minimize this uncertainty,
basecalling algorithms use statistical models, heuristics, or more recently,
deep learning. By modeling the errors and biases, and sometimes incorpor-
ating other related prior information, variant callers that use statistical
models significantly reduces the probability of miscalling variants. Methods
that are based on the heuristic approach, on the other hand, call variants
based on a number of heuristic factors, such as minimum read depth, base
quality, and allele frequency. Deep learning is a subfield of machine learning
that uses artificial neural networks (ANNs) to learn from training data with
known truth, to make predictions or classifications from new unknown data.
To call variants from NGS data using deep learning, pre-​labeled variants are
first used as training data to build a model for subsequent variant calling.
Among the algorithms that use these three different approaches, those based
on statistical models are currently more widely used than those based on
218 Next-Generation Sequencing Data Analysis

C
T
T
C
T
C
C

C
FIGURE 10.2
The variant calling process is usually affected by various factors. In this illustration, a number
of reads are aligned against a reference sequence (bottom). At the illustrated site, the reference
sequence has a C while the reads have C and T. Depending on the factors mentioned in the text
and prior information, this site can be called as heterozygous (C/T), or no variation (C/C) if
the T’s are treated as errors. It is also possible to be called as a homozygous T/T, if the C’s are
regarded as errors.

heuristics and deep learning. It should be noted, however, that statistical


models are usually based on certain assumptions. Under circumstances
when the assumptions are violated, the heuristic and deep learning-​based
methods can be more robust.
Among the variant callers that are based on statistical models, GATK [4] is
currently among the most widely used. GATK mostly uses HaplotypeCaller
to call germline SNVs and short indels. HaplotypeCaller, as the name
suggests, considers the linkage between nearby variants and calls SNPs and
indels simultaneously through local de novo haplotype assembly. More spe-
cifically, the variant calling process used by HaplotypeCaller is composed
of four main steps: (1) identification of active genomic regions that most
likely harbor variants based on evidence from reads; (2) determination of
haplotypes for each active region via reassembly of the region using a De
Bruijn-​like graph, followed by realignment of the haplotypes against the ref-
erence to identify potential variant sites; (3) calculation of the likelihoods of
the haplotypes through aligning each read against each haplotype using a
Pair-​Hidden Markov Model (PairHMM); and (4) application of Bayes’ rule to
find the most likely genotypes in the sample. This variant calling process is
highly accurate, but it is also computationally intensive and relatively slow.
BCFtools [5] uses a similar genotype likelihood model for variant calling,
which is achieved in two steps, namely “bcftools mpileup” and “bcftools
call.” In the mpileup step, it collects summary information from input BAM
files and computes the likelihoods of possible genotypes, which are stored in
BCF files (to be detailed next). The subsequent call step uses the likelihood
information in the BCF files to conduct variant calling.
Whole Genome/Exome Sequencing 219

Besides GATK and BCFtools, other model-​based variant callers include


Strelka2 [6], freebayes [7], and 16GT [8]. A commonly used heuristics-​based
variant caller is VarScan2 [9], which works more robustly on data confounded
by factors such as extreme read depth, pooled samples, and contaminated or
impure samples. Many of these tools, such as GATK, BCFtools, Strelka2, and
VarScan2, work for both single-​and multiple-​sample data. Multiple-​sample
analysis usually has increased detection power than single-​sample analysis
[10], because with multiple samples it is more likely to call a variant when
more than one sample shows the same variation.
Google’s open-​source DeepVariant tool was the first attempt to use deep
learning for variant calling. It is based on the use of convolutional neural
network (CNN), a class of ANN that is often applied to image classification
problems [11]. With the use of CNN, the variant calling process is converted
into an image classification process [12]. With the increased use of long reads
from the ONT and PacBio platforms, many of the variant callers specific-
ally developed for long reads are based on the use of deep learning. These
tools include Clair3 [13], Longshot [14], NanoCaller [15], and Medaka [16].
NanoCaller, for example, combines long-​range haplotype information and
deep CNN in an effort to improve variant calling accuracy. DeepVariant
can also be used on long reads besides short reads, when coupled with
algorithms such as PEPPER that uses a recurrent neural network to find
variant candidates for subsequent variant calling by DeepVariant [17]. Based
on currently available benchmarking studies [18–​20], DeepVariant has been
shown to have similar or even better performance than the often used GATK
pipeline.

10.2.2 Somatic Mutation Detection


Most of the currently available variant calling methods are designed to
identify germline variations that can be passed from generation to gener-
ation. While these variants are major detection targets, somatic mutations
(Figure 10.3) also play important roles in many diseases such as cancer. To
identify such acquired somatic mutations, some of the germline variant
callers mentioned above also provide utilities or options designed for calling
somatic mutations, such as GATK Mutect2 [21], Strelka2, and VarScan2. Tools
specifically designed for somatic mutation detection include SomaticSniper
[22], VarDict [23], NeuSomatic [24], JointSNVmix [25], and Lancet [26].
Mechanistically, while some of these algorithms (such as Mutect2 and
VarScan2) carry out mutation calling on each of the contrasting samples (e.g.,
normal vs. cancer tissues carrying somatic mutations from the same patient)
separately against a reference genome, others (such as Strelka2, JointSNVmix,
and SomaticSniper) directly compare the contrasting samples. In the former
approach, sequence reads generated from contrasting samples are independ-
ently aligned to and variants called against a reference genome. The called
220 Next-Generation Sequencing Data Analysis

FIGURE 10.3
Detection of somatic mutations vs. germline variations. In this example, sequence reads from
normal and tumor tissues are aligned to the reference genome (shown at the top in green). The
allelic counts, i.e., the number of matches (aN and aT) and depth of reads (dN and dT), at each base
position are shown. The blue sites show germline positions, while the red shows a position where
a somatic mutation occurred in some tumor cells. Also shown at the bottom are the predicted
genotypes for the normal and tumor tissues. (Modified from Roth A. et al. JointSNVMix: a
probabilistic model for accurate detection of somatic mutations in normal/​ tumour paired
next-​generation sequencing data. Bioinformatics, 2012, 28 (7): 907–​13, by permission of Oxford
University Press.)

variants in the contrasting samples are then compared to each other to locate
somatic mutations in the cancer tissue. In the latter approach, the samples
are directly compared to each other using statistical tests on the basis of joint
probability. NeuSomatic represents the first attempt to use deep learning
(CNN) for somatic variant detection. The currently developed Mutect3 also
uses machine learning in an effort to improve somatic mutation detection
accuracy. To help evaluate the performance of these various somatic variant
callers, several benchmarking studies [27–​30] are available showing that
Mutect2 and Strelka2 are among the top performers so far.
Whole Genome/Exome Sequencing 221

10.2.3 Variant Calling from RNA Sequencing Data


While variant calling is mostly carried out from DNA sequencing data,
RNA-​ seq can also be used to call variants from transcriptionally active
regions of the genome. RNA-​seq-​based variant calling is more challenging
due to the inherent heterogeneity in the abundance of reads transcribed
from different regions, differential splicing of exons, allele-​specific expres-
sion, RNA editing, etc. Variant calling from RNA-​seq data offers certain
advantages, however, as it does not incur additional cost beyond collecting
the original transcriptomic data, and it directly interrogates transcription-
ally active regions of the genome. In addition, RNA-​seq-​based variant dis-
covery can be used to validate variants called from whole-​genome or –​exome
sequencing. Methods for RNA-​seq-​based variant calling are still limited.
Currently available tools/​pipelines include the GATK best practices work-
flow for RNA-​seq short variant calling [31], SNPiR [32], eSNV-​Detect [33],
SNVMix [34], and Opossum-​Platypus [35]. Most of these tools are based on
the use of variant callers developed for germline DNA sequencing, such as
the GATK HaplotypeCaller or BCFtools. The often used GATK best practices
workflow for RNA-​seq data, for example, centers around HaplotypeCaller
and comprises multiple steps including: 1) alignment of raw RNA-​seq reads
to the reference genome using STAR; 2) cleanup of data through identifica-
tion and removal of duplicate reads; 3) reformatting of alignments that span
intronic regions as a preparatory step for variant calling; 4) recalibration of
base quality scores to improve variant calling accuracy; 5) variant calling
using HaplotypeCaller; and 6) variant filtering to produce the final list of
variants. SNVMix, on the other hand, employs a probabilistic binomial mix-
ture model to call variants from pre-​mapped RNA-​seq reads.

10.2.4 Variant Call Format (VCF)


VCF is a text-​based standard file format for storing sequence variations,
including SNVs, short indels, and SVs (to be detailed next) [36]. This format
is designed to be scalable to encompass millions of sites from thousands of
samples. Originally developed for the 1000 Genomes Project, it is designed for
fast data retrieval. Besides reporting variants and their genomic positions, it
offers fields to store additional information such as variant call quality score,
and allows users to add their own custom tags to describe new sequence
variations. BCF is the binary version of VCF, providing speed and efficiency
through compression of the variant data.
Figure 10.4 provides an example of the VCF format. It contains meta-​
information lines at the front, a header line, and data lines each of which
describes a variant position. The metainfo lines start with “##” and describe
related analysis information, such as species, file date, and assembly version.
In addition, abbreviations used in the user definable data columns are also
defined in the metainfo lines. The subsequent header line lists the names of
newgenrtpdf
222
##fileformat=VCFv4.3
##fileDate=20090805
##source=myImputationProgramV3.1
##reference=file:///seq/references/1000GenomesPilot-NCBI36.fasta
##contig=<ID=20,length=62435964,assembly=B36,md5=f126cdf8a6e0c7f379d618ff66beb2da,species="Homo sapiens",taxonomy=x>
##phasing=partial
##INFO=<ID=NS,Number=1,Type=Integer,Description="Number of Samples With Data">
##INFO=<ID=DP,Number=1,Type=Integer,Description="Total Depth">
##INFO=<ID=AF,Number=A,Type=Float,Description="Allele Frequency">
##INFO=<ID=AA,Number=1,Type=String,Description="Ancestral Allele">
##INFO=<ID=DB,Number=0,Type=Flag,Description="dbSNP membership, build 129">
##INFO=<ID=H2,Number=0,Type=Flag,Description="HapMap2 membership">
##FILTER=<ID=q10,Description="Quality below 10">

Next-Generation Sequencing Data Analysis


##FILTER=<ID=s50,Description="Less than 50% of samples have data">
##FORMAT=<ID=GT,Number=1,Type=String,Description="Genotype">
##FORMAT=<ID=GQ,Number=1,Type=Integer,Description="Genotype Quality">
##FORMAT=<ID=DP,Number=1,Type=Integer,Description="Read Depth">
##FORMAT=<ID=HQ,Number=2,Type=Integer,Description="Haplotype Quality">
#CHROM POS ID REF ALT QUAL FILTER INFO FORMAT NA00001 NA00002 NA00003
20 14370 rs6054257 G A 29 PASS NS=3;DP=14;AF=0.5;DB;H2 GT:GQ:DP:HQ 0|0:48:1:51,51 1|0:48:8:51,51 1/1:43:5:.,.
20 17330 . T A 3 q10 NS=3;DP=11;AF=0.017 GT:GQ:DP:HQ 0|0:49:3:58,50 0|1:3:5:65,3 0/0:41:3
20 1110696 rs6040355 A G,T 67 PASS NS=2;DP=10;AF=0.333,0.667;AA=T;DB GT:GQ:DP:HQ 1|2:21:6:23,27 2|1:2:0:18,2 2/2:35:4
20 1230237 . T . 47 PASS NS=3;DP=13;AA=T GT:GQ:DP:HQ 0|0:54:7:56,60 0|0:48:4:51,51 0/0:61:2
20 1234567 microsat1 GTC G,GTCT 50 PASS NS=3;DP=9;AA=G GT:GQ:DP 0/1:35:4 0/2:17:2 1/1:40:3
FIGURE 10.4
The VCF format (version 4.3). The format is currently managed by the Large Scale Genomics Work Stream, part of the Global Alliance for Genomics
& Health (GA4GH). BCF is the binary counterpart of VCF. (From http://​samto​ols.git​hub.io/​hts-​specs/.​)
Whole Genome/Exome Sequencing 223

TABLE 10.1
Mandatory Fields in a VCF File
Col Field Type Description

1 #CHROM String Chromosome number


2 POS Integer Start position of the variation
3 ID String Database identifier
4 REF String Reference allele
5 ALT String Alternate allele(s)
6 QUAL Numeric Quality score (Phred-​style)
7 FILTER String Filter status
8 INFO String User extensible information

the 8 mandatory columns (Table 10.1). In the QUAL column, a Phred-​like


quality score for the alternative allele (ALT) call is given (e.g., a QUAL value
of 30 means the probability of the ALT call being wrong is 0.001). In the
FILTER column, “PASS” means this position has passed all filters, while a
value of “q10” as shown in Figure 10.4 indicates that the variant call quality
at this site is below 10. The data lines, containing variant calls for a list of gen-
omic positions, make the body of a VCF file.
VCF/​BCF files can be parsed, manipulated, and visualized using tools
such BCFtools and vcfR [37]. BCFtools, beyond its capability to call variants,
offers a set of utilities for VCF/​BCF files on a long range of operations, such
as file format conversion, summarizing variant statistics, variant filtering,
sorting, concatenation or merging of multiple files, finding how variants in
multiple files intersect, detection of sample swaps and contamination, etc.
Besides built-​in commands for such operations, it also supports the use of
plugins for specific single-​purpose tasks. Examples of such plugins and tasks
are frameshifts to annotate frameshift indels, guess-​ploidy to determine sample
sex, split-​vep to query and extract from variants’ VEP annotations (see Section
10.4 for VEP variant annotation), trio-​dnm2 to determine de novo mutations in
parents-​offspring trios, and gvcfz to compress gVCF (or genomic VCF) files.
The gVCF format is an extended version of VCF with the same format speci-
fication, but it also contains information for the rest of non-​variant genomic
regions with confidence estimates that they match the reference genome. The
goal to include all sites of the genome in a gVCF is to facilitate joint geno-
typing of a cohort of samples when needed.

10.2.5 Evaluating VCF Results


SNVs and indels reported in VCF files need to be evaluated to identify false
positives. Visualization of called variants and supporting reads in a genome
browser, such as IGV or Savant, provides an initial examination of the variant
call result. Further evaluation should be based on criteria such as deviation
from Hardy–​ Weinberg equilibrium, systematic call quality difference
224 Next-Generation Sequencing Data Analysis

Predicted
Positive Negative

Positive
True Positive False Negative
(TP) (FN)

Actual
Negative False Positive True Negative
(FP) (TN)

FIGURE 10.5
Contingency table for variant calling.

between major and minor alleles, extreme depth of coverage, or strand bias.
The ratio of transitions and transversions (Ti/​Tv) is an additional indicator
of variant call specificity and quality. The theoretical ratio of Ti/​Tv is 0.5,
because purely from the point of statistical probability the chance of produ-
cing transitions is half that of transversions. However, due to biochemical
mechanisms involved in these nucleotide substitution processes, the
frequency of having transitions is higher than that of transversions. Based
on existing NGS data from multiple species, the expected values of Ti/​Tv
for whole genome and exome datasets are usually in the ranges of 2.0–​2.1,
3.0–​3.5, respectively [38]. Variants that do not pass these QC criteria are then
filtered out. Besides such filtering using preset criteria, low quality variants
may also be identified for removal using machine learning approaches such
as ForestQC [39] and VQSR (part of GATK).
As different variant callers employ different approaches, the variants they
identify usually only partially overlap. It is advisable, therefore, to examine
closely on the specifics of an experiment to decide on more appropriate
variant caller(s). If more than one method is used, it is advisable to compare
their outputs and analyze how they intersect. Use of convergent variants
is an effective way to reduce rates of miscalled variants. Alternatively,
ensemble methods such as VariantMetaCaller [40] and BAYSIC [41] can also
be used.
To further compare results from multiple variant callers side-​by-​side, pre-
cision and recall are key metrics often used to measure the performance of
variant callers. Precision refers to the ability to not detect false positives,
while recall to not detect false negatives (Figure 10.5). Mathematically, preci-
sion and recall are calculated as

TP
Precision =
TP + FP ( i.e., all predicted positives)
Whole Genome/Exome Sequencing 225

TP
Recall =
TP + FN ( i.e., all actual positives)

To combine precision and recall into a single metric to simplify evaluation


of variant callers, F1 score is often used. Mathematically, F1 is the harmonic
mean of the two, calculated as

Precision × Recall
F1 = 2 ×
Precision + Recall

By using the harmonic mean instead of the arithmetic mean, F1 provides a


more balanced way to measure variant calling performance. For example, if
either precision or recall is low, the F1 score is low. To have a high F1 score,
both precision and recall need to be high. Utilities such as RTG Tools [42] can
help compare results from multiple callers.

10.3 Structural Variant (SV) Calling


10.3.1 Short-Read-​Based SV Calling
As covered in Chapter 2, structural variants in the genome involve insertions,
deletions, duplications, inversions, and translocations of sequences at least
50 bp in size. SVs are largely the basis of genome evolution and diversifi-
cation, and produce more genomic differences than SNVs between individ-
uals [43]. Earlier experimental methods on the detection of SVs were mostly
based on comparative genome hybridization and SNP whole genome arrays.
The advent of NGS, especially the use of paired-​end short reads and more
recently long reads, has greatly pushed SV detection forward. As illustrated
in Figure 10.6, the basic approaches to detect SVs using short reads are based
on: (1) the change in read depth (RD) as compared to the rest of the genome;
(2) the use of paired-​reads (PR) to detect unexpected change in orientation,
distance between them, or their localization to different chromosomes; (3) the
existence of split-​reads (SR) that span disjoint regions of the genome; and
(4) the deviation of de novo assembled (AS) sequence from the reference
sequence.
Among these approaches, the RD approach is to be detailed in Section
10.3.3. Figure 10.7 illustrates the main steps in the PR approach. The first
step is to separate read pairs into concordant or discordant groups, defined
by the distance between a read pair matching or deviating from the expected
distance based on the reference genome. The discordant read pairs are then
assembled into different clusters based on the genomic region they cover
to generate candidate SV calling regions. In the last step, the candidate SV
226 Next-Generation Sequencing Data Analysis

FIGURE 10.6
Four approaches for SV detection. (From Escaramís, G. & Docampo, E. A decade of structural
variants: description, history and methods to detect structural variation. Briefings in Functional
Genomics, 2015, 14 (5): 305–​14, by permission of Oxford University Press.)

FIGURE 10.7
General steps of calling SVs using paired-​end reads. (Used with permission from Whelan,
Christopher, “Detecting and Analyzing Genomic Structural Variation Using Distributed
Computing” (2014). Scholar Archive. Paper 3482.)

clusters are filtered based on statistical assessment so that only clusters that
are covered by multiple read pairs are reported as SVs. The boundaries of
possible break points in the region are also identified in this step (indicated
by the shaded area in Figure 10.7, panel d). Among currently available SV
detection algorithms, PEMer (or Paired-​End Mapper) [44], BreakDancer [45],
Whole Genome/Exome Sequencing 227

SVDetect [46], and 1-​2-​3-​SV [47] apply this paired-​reads based approach.
Pindel [48] provides an example for the SR approach. It first searches for
read pairs in which one read aligns to the reference genome but the other
does not. Based on the assumption that the second read contains a break-
point, it uses the aligned read as anchor to scan the surrounding regions for
split mapping of the second read. While it can locate breakpoints at single
base resolution, this approach is computationally expensive because of the
challenge associated with aligning read sub-​sequences to different genomic
regions with gaps in between. Cortex [49] and AsmVar [50] are examples of
the DS approach. In this approach, the genome is first assembled from reads,
and subsequently SVs are called through alignment and statistical analysis of
the de novo genome assembly against the reference.
To improve detection accuracy, many currently available SV detection
algorithms use a combination of these approaches. For example, DELLY [51],
Meerkat [52], SoftSV [53], and Wham [54] combine PR and SR, while GASV/​
GASVPro [55, 56], Genome STRiP [57], and inGAP-​sv [58] combine RD and
PR. HYDRA [59] is an example of combining RD and AS. As examples of
combining three approaches, MANTA [60], GRIDSS [61], SvABA [62], and
CREST [63] combine PR, SR, and AS. LUMPY [64] and TIDDIT [65], on the
other hand, combine RD, PR, and SR.

10.3.2 Long-Read-​Based SV Calling
SV detection from the use of short reads has high miscalling rates because
of the limitations of short-read sequencing. Long-read technologies such as
PacBio and Oxford Nanopore sequencing overcome such inherent limitations
caused by short read length. Mechanistically, long-read-​based SV callers
are mostly built on the use of the SR and/​or AS approaches. These callers
include pbsv [66], Sniffles [67], Phased Assembly Variant (PAV) [68], MELT
[69], NanoVar [70], NanoSV [71], PALMER [72], SVIM [73], and Picky [74].
Some callers, such as Dysgu [75], are developed to use both long and short
reads. Besides long reads generated from long-read sequencers, synthetic
long reads obtained using technologies such as linked-​read sequencing [76–​
79] can also be used for SV detection by deploying tools such as Long Ranger,
an open-​source pipeline developed by 10× Genomics [80].

10.3.3 CNV Detection
CNVs, caused by duplications, insertions, or deletions, are an important
subtype of structural variation. Among the four basic approaches outlined
in Section 10.3.1, CNV detection algorithms are often based on RD. These
algorithms are based on the assumption that the number of reads obtained
from a region is proportional to its copy number in the genome. If a gen-
omic segment is repeated multiple times, for example, a significantly higher
number of reads will be observed from the segment compared to other
228 Next-Generation Sequencing Data Analysis

non-​repeated regions. If a segment is deleted, on the other hand, there will be


no read coverage for it (see Figure 10.6, panel A). These RD-​based algorithms
include CNVnator [81], CNV-​ seq [82], CNVkit [83], cn.MOPS (Copy
Number estimation by a Mixture of PoissonS) [84], CNAseg [85], ERDS [86],
RDXplorer (Read Depth eXplorer) [87], Control-​FREEC [88], mrFAST [89],
SegSeq [90], readDepth [91], Canvas [92], and iCopyDAV [93]. Among these
algorithms, CNVnator, built on the use of a mean-​shift approach originally
designed for image processing, is one of the most commonly used. As factors
such as GC content may also affect local read density, a normalization step
is often conducted when deploying these algorithms to account for these
compounding factors. In studies that involve comparison of samples from
the same genetic background, e.g., a diseased tissue vs. a healthy tissue from
the same patient, these compounding factors are often cancelled out.

10.3.4 Integrated SV Analysis
The different software tools introduced above are based on different algo-
rithmic design, and as a result show varying performance for detecting par-
ticular types (or aspects) of SVs [94, 95]. In order to improve call performance
for the full range of SVs, there have been efforts to take an integrated approach
towards comprehensive SV calling using the different but often complemen-
tary tools. SVMerge, being one of these efforts, integrates SV calling results
from different callers [96]. It first feeds BAM files into a number of SV callers
including those introduced above to generate BED files, and then the SV calls
in the BED files are merged. After computational validation and breakpoint
refinement by local de novo alignment, a final list of SVs is generated. Other
efforts that take a similarly integrated approach include Parliament2 [97],
FusorSV [98], SURVIVOR [99], MetaSV [100], and CNVer [101].

10.4 Annotation of Called Variants


To gain biological insights from identified SNVs, indels, or SVs, annota-
tion of the variants is needed. For example, if an SNV is annotated to be
nonsynonymous in a gene, it may impair protein function if the affected
amino acid is located within the active site of the protein. Through exam-
ination of their annotations, called variants can be filtered and prioritized
for more in-​depth analysis. Because of the large number of variants usually
called from an experiment, an automatic pipeline is usually preferred. To
meet this demand a number of variant annotation tools have been developed.
ANNOVAR [102] is one such tool among the most widely used. It takes SNVs,
indels, and CNVs as input, and as output, it reports their functional impacts
Whole Genome/Exome Sequencing 229

and provides significance scores to help with filtering and prioritization.


Its TABLE_​ANNOVAR script can quickly turn a variant list into an Excel-​
compatible file containing many annotation fields that can help researcher
evaluate the function importance of the variants. ANNOVAR offers flexi-
bility and extensibility, e.g., it can identify variants located in conserved gen-
omic regions, or find variants that overlap with those from the 1000 genomes
project or dbSNP. Other widely used variant annotation tools include SnpEff
[103], VEP (Variant Effect Predictor) [104], UCSC Genome Browser’s Variant
Annotation Integrator [105], and SeattleSeq [106].
On the back end of these annotation tools are various types of annotation
databases. These databases provide reference information on what genes
or regions of the genome they are at (e.g., transcriptional factor binding
sites, DNase I hypersensitivity sites, or highly conserved regions), what
functional consequence(s) they might cause based on known phenotype or
computational inference, whether an identified variant has been observed
before, how frequently they are observed in different populations if previ-
ously known, whether there is evidence for their connection with a human
disease (especially relevant for human patient samples), etc. Annotational
information from these databases is essential for filtering out non-​target
variants, and ranking/​prioritizing the remaining variants for reporting pur-
pose and further analyses. Many of these databases, as well as the detailed
steps of variant filtering, ranking, and prioritizing, are covered in detail in
the next chapter.

References
1. Acuna-​Hidalgo R, Veltman JA, Hoischen A. New insights into the gener-
ation and role of de novo mutations in health and disease. Genome Biol 2016,
17(1):241.
2. Miller MB, Reed HC, Walsh CA. Brain Somatic Mutation in Aging and
Alzheimer’s Disease. Annu Rev Genomics Hum Genet 2021, 22:239–​256.
3. Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells.
Science 2015, 349(6255):1483–​1489.
4. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky
A, Garimella K, Altshuler D, Gabriel S, Daly M et al. The Genome Analysis
Toolkit: a MapReduce framework for analyzing next-​generation DNA sequen-
cing data. Genome Res 2010, 20(9):1297–​1303.
5. Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V, Pollard MO, Whitwham
A, Keane T, McCarthy SA, Davies RM et al. Twelve years of SAMtools and
BCFtools. GigaScience 2021, 10(2).
6. Kim S, Scheffler K, Halpern AL, Bekritsky MA, Noh E, Kallberg M, Chen X,
Kim Y, Beyter D, Krusche P et al. Strelka2: fast and accurate calling of germline
and somatic variants. Nat Methods 2018, 15(8):591–​594.
230 Next-Generation Sequencing Data Analysis

7. Garrison E, Marth G. Haplotype-​ based variant detection from short-​ read


sequencing. arXiv:12073907, 2012.
8. Luo R, Schatz MC, Salzberg SL. 16GT: a fast and sensitive variant caller using
a 16-​genotype probabilistic model. GigaScience 2017, 6(7):1–​4.
9. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis
ER, Ding L, Wilson RK. VarScan 2: somatic mutation and copy number alteration
discovery in cancer by exome sequencing. Genome Res 2012, 22(3):568–​576.
10. Liu J, Shen Q, Bao H. Comparison of seven SNP calling pipelines for the next-​
generation sequencing data of chickens. PLoS One 2022, 17(1):e0262574.
11. Valueva MV, Nagornov N, Lyakhov PA, Valuev GV, Chervyakov NI.
Application of the residue number system to reduce hardware costs of the
convolutional neural network implementation. Math Comput Simul 2020,
177:232–​243.
12. Poplin R, Chang PC, Alexander D, Schwartz S, Colthurst T, Ku A, Newburger
D, Dijamco J, Nguyen N, Afshar PT et al. A universal SNP and small-​indel
variant caller using deep neural networks. Nat Biotechnol 2018, 36(10):983–​987.
13. Zheng Z, Li S, Su J, Leung AW-​S, Lam T-​W, Luo R. Symphonizing pileup and
full-​alignment for deep learning-​based long-​read variant calling. Nat Comput
Sci 2022, 2(12):797–​803.
14. Edge P, Bansal V. Longshot enables accurate variant calling in diploid genomes
from single-​molecule long read sequencing. Nat Commun 2019, 10(1):4660.
15. Ahsan MU, Liu Q, Fang L, Wang K. NanoCaller for accurate detection of
SNPs and indels in difficult-​to-​map regions from long-​read sequencing by
haplotype-​aware deep neural networks. Genome Biol 2021, 22(1):261.
16. (https://​git​hub.com/​nanop​oret​ech/​med​aka)
17. Shafin K, Pesout T, Chang PC, Nattestad M, Kolesnikov A, Goel S, Baid G,
Kolmogorov M, Eizenga JM, Miga KH et al. Haplotype-​aware variant calling
with PEPPER-​Margin-​DeepVariant enables high accuracy in nanopore long-​
reads. Nat Methods 2021, 18(11):1322–​1332.
18. Supernat A, Vidarsson OV, Steen VM, Stokowy T. Comparison of three variant
callers for human whole genome sequencing. Sci Rep 2018, 8(1):17851.
19. Pei S, Liu T, Ren X, Li W, Chen C, Xie Z. Benchmarking variant callers in next-​
generation and third-​generation sequencing analysis. Brief Bioinform 2021,
22(3):bbaa148.
20. Lin YL, Chang PC, Hsu C, Hung MZ, Chien YH, Hwu WL, Lai F, Lee NC.
Comparison of GATK and DeepVariant by trio sequencing. Sci Rep 2022,
12(1):1809.
21. Benjamin D, Sato T, Cibulskis K, Getz G, Stewart C, Lichtenstein L. Calling
somatic SNVs and indels with Mutect2. bioRxiv 2019, doi: https://​doi.org/​
10.1101/​861​054
22. Larson DE, Harris CC, Chen K, Koboldt DC, Abbott TE, Dooling DJ, Ley
TJ, Mardis ER, Wilson RK, Ding L. SomaticSniper: identification of som-
atic point mutations in whole genome sequencing data. Bioinformatics 2012,
28(3):311–​317.
23. Lai Z, Markovets A, Ahdesmaki M, Chapman B, Hofmann O, McEwen R,
Johnson J, Dougherty B, Barrett JC, Dry JR. VarDict: a novel and versatile
variant caller for next-​generation sequencing in cancer research. Nucleic Acids
Res 2016, 44(11):e108.
Whole Genome/Exome Sequencing 231

24. Sahraeian SME, Liu R, Lau B, Podesta K, Mohiyuddin M, Lam HYK. Deep
convolutional neural networks for accurate somatic mutation detection. Nat
Commun 2019, 10(1):1041.
25. Roth A, Ding J, Morin R, Crisan A, Ha G, Giuliany R, Bashashati A, Hirst M,
Turashvili G, Oloumi A et al. JointSNVMix: a probabilistic model for accurate
detection of somatic mutations in normal/​tumour paired next-​generation
sequencing data. Bioinformatics 2012, 28(7):907–​913.
26. Narzisi G, Corvelo A, Arora K, Bergmann EA, Shah M, Musunuri R, Emde
AK, Robine N, Vacic V, Zody MC. Genome-​wide somatic variant calling using
localized colored de Bruijn graphs. Commun Biol 2018, 1:20.
27. Cai L, Yuan W, Zhang Z, He L, Chou KC. In-​depth comparison of somatic
point mutation callers based on different tumor next-​generation sequencing
depth data. Sci Rep 2016, 6:36540.
28. Kroigard AB, Thomassen M, Laenkholm AV, Kruse TA, Larsen MJ. Evaluation
of nine somatic variant callers for detection of somatic mutations in exome
and targeted deep sequencing data. PLoS One 2016, 11(3):e0151664.
29. Chen Z, Yuan Y, Chen X, Chen J, Lin S, Li X, Du H. Systematic comparison of
somatic variant calling performance among different sequencing depth and
mutation frequency. Sci Rep 2020, 10(1):3501.
30. Zhao S, Agafonov O, Azab A, Stokowy T, Hovig E. Accuracy and efficiency
of germline variant calling pipelines for human genome data. Sci Rep 2020,
10(1):20222.
31. GATK Best Practices Workflow for RNAseq Short Variant Discovery (SNPs
+​Indels) (https://​gatk.bro​adin​stit​ute.org/​hc/​en-​us/​artic​les/​36003​5531​192-​
RNA​seq-​short-​vari​ant-​discov​ery-​SNPs-​Ind​els-​)
32. Piskol R, Ramaswami G, Li JB. Reliable identification of genomic variants
from RNA-​seq data. Am J Hum Genet 2013, 93(4):641–​651.
33. Tang X, Baheti S, Shameer K, Thompson KJ, Wills Q, Niu N, Holcomb IN,
Boutet SC, Ramakrishnan R, Kachergus JM et al. The eSNV-​detect: a computa-
tional system to identify expressed single nucleotide variants from transcrip-
tome sequencing data. Nucleic Acids Res 2014, 42(22):e172.
34. Goya R, Sun MG, Morin RD, Leung G, Ha G, Wiegand KC, Senz J, Crisan A,
Marra MA, Hirst M et al. SNVMix: predicting single nucleotide variants from
next-​generation sequencing of tumors. Bioinformatics 2010, 26(6):730–​736.
35. Oikkonen L, Lise S. Making the most of RNA-​seq: Pre-​processing sequen-
cing data with Opossum for reliable SNP variant detection. Wellcome Open Res
2017, 2:6.
36. Danecek P, Auton A, Abecasis G, Albers CA, Banks E, DePristo MA, Handsaker
RE, Lunter G, Marth GT, Sherry ST et al. The variant call format and VCFtools.
Bioinformatics 2011, 27(15):2156–​2158.
37. Knaus BJ, Grunwald NJ. vcfr: a package to manipulate and visualize variant
call format data in R. Mol Ecol Resour 2017, 17(1):44–​53.
38. Freudenberg-​Hua Y, Freudenberg J, Kluck N, Cichon S, Propping P, Nothen
MM. Single nucleotide variation analysis in 65 candidate genes for CNS
disorders in a representative sample of the European population. Genome Res
2003, 13(10):2271–​2276.
39. Li J, Jew B, Zhan L, Hwang S, Coppola G, Freimer NB, Sul JH. ForestQC: Quality
control on genetic variants from next-​ generation sequencing data using
random forest. PLoS Comput Biol 2019, 15(12):e1007556.
232 Next-Generation Sequencing Data Analysis

40. Gezsi A, Bolgar B, Marx P, Sarkozy P, Szalai C, Antal P. VariantMetaCaller:


automated fusion of variant calling pipelines for quantitative, precision-​based
filtering. BMC Genomics 2015, 16:875.
41. Cantarel BL, Weaver D, McNeill N, Zhang J, Mackey AJ, Reese J. BAYSIC: a
Bayesian method for combining sets of genome variants with improved speci-
ficity and sensitivity. BMC Bioinformatics 2014, 15:104.
42. RTG Tools: Utilities for accurate VCF comparison and manipulation (https://​
git​hub.com/​RealT​imeG​enom​ics/​rtg-​tools)
43. Sudmant PH, Rausch T, Gardner EJ, Handsaker RE, Abyzov A, Huddleston J,
Zhang Y, Ye K, Jun G, Fritz MH et al. An integrated map of structural variation
in 2,504 human genomes. Nature 2015, 526(7571):75–​81.
44. Korbel JO, Abyzov A, Mu XJ, Carriero N, Cayting P, Zhang Z, Snyder M,
Gerstein MB. PEMer: a computational framework with simulation-​ based
error models for inferring genomic structural variants from massive paired-​
end sequencing data. Genome Biol 2009, 10(2):R23.
45. Chen K, Wallis JW, McLellan MD, Larson DE, Kalicki JM, Pohl CS, McGrath
SD, Wendl MC, Zhang Q, Locke DP et al. BreakDancer: an algorithm for
high-​resolution mapping of genomic structural variation. Nat Methods 2009,
6(9):677–​681.
46. Zeitouni B, Boeva V, Janoueix-​ Lerosey I, Loeillet S, Legoix-​ ne P, Nicolas
A, Delattre O, Barillot E. SVDetect: a tool to identify genomic structural
variations from paired-​ end and mate-​ pair sequencing data. Bioinformatics
2010, 26(15):1895–​1896.
47. 1-​2-​3-​SV (https://​git​hub.com/​Vit​yay/​1-​2-​3-​SV)
48. Ye K, Guo L, Yang X, Lamijer EW, Raine K, Ning Z. Split-​read indel and struc-
tural variant calling using PINDEL. Methods Mol Biol 2018, 1833:95–​105.
49. Iqbal Z, Caccamo M, Turner I, Flicek P, McVean G. De novo assembly and
genotyping of variants using colored de Bruijn graphs. Nat Genet 2012,
44(2):226–​232.
50. Liu S, Huang S, Rao J, Ye W, Genome Denmark Consortium II, Krogh A, Wang
J. Discovery, genotyping and characterization of structural variation and novel
sequence at single nucleotide resolution from de novo genome assemblies on
a population scale. GigaScience 2015, 4 :64.
51. Rausch T, Zichner T, Schlattl A, Stutz AM, Benes V, Korbel JO. DELLY: struc-
tural variant discovery by integrated paired-​ end and split-​
read analysis.
Bioinformatics 2012, 28(18):i333–​i339.
52. Yang L, Luquette LJ, Gehlenborg N, Xi R, Haseley PS, Hsieh CH, Zhang C,
Ren X, Protopopov A, Chin L et al. Diverse mechanisms of somatic structural
variations in human cancer genomes. Cell 2013, 153(4):919–​929.
53. Bartenhagen C, Dugas M. Robust and exact structural variation detection
with paired-​end and soft-​clipped alignments: SoftSV compared with eight
algorithms. Brief Bioinform 2016, 17(1):51–​62.
54. Kronenberg ZN, Osborne EJ, Cone KR, Kennedy BJ, Domyan ET, Shapiro MD,
Elde NC, Yandell M. Wham: Identifying structural variants of biological con-
sequence. PLoS Comput Biol 2015, 11(12):e1004572.
55. Sindi S, Helman E, Bashir A, Raphael BJ. A geometric approach for clas-
sification and comparison of structural variants. Bioinformatics 2009,
25(12):i222–​230.
Whole Genome/Exome Sequencing 233

56. Sindi SS, Onal S, Peng LC, Wu HT, Raphael BJ. An integrative probabilistic
model for identification of structural variation in sequencing data. Genome Biol
2012, 13(3):R22.
57. Handsaker RE, Van Doren V, Berman JR, Genovese G, Kashin S, Boettger LM,
McCarroll SA. Large multiallelic copy number variations in humans. Nat
Genet 2015, 47(3):296–​303.
58. Qi J, Zhao F. inGAP-​sv: a novel scheme to identify and visualize structural
variation from paired end mapping data. Nucleic Acids Res 2011, 39(Web
Server issue):W567–​575.
59. Quinlan AR, Clark RA, Sokolova S, Leibowitz ML, Zhang Y, Hurles ME,
Mell JC, Hall IM. Genome-​wide mapping and assembly of structural variant
breakpoints in the mouse genome. Genome Res 2010, 20(5):623–​635.
60. Chen X, Schulz-​Trieglaff O, Shaw R, Barnes B, Schlesinger F, Kallberg M, Cox
AJ, Kruglyak S, Saunders CT. Manta: rapid detection of structural variants
and indels for germline and cancer sequencing applications. Bioinformatics
2016, 32(8):1220–​1222.
61. Cameron DL, Schroder J, Penington JS, Do H, Molania R, Dobrovic A,
Speed TP, Papenfuss AT. GRIDSS: sensitive and specific genomic rearrange-
ment detection using positional de Bruijn graph assembly. Genome Res 2017,
27(12):2050–​2060.
62. Wala JA, Bandopadhayay P, Greenwald NF, O’Rourke R, Sharpe T, Stewart
C, Schumacher S, Li Y, Weischenfeldt J, Yao X et al. SvABA: genome-​wide
detection of structural variants and indels by local assembly. Genome Res 2018,
28(4):581–​591.
63. Wang J, Mullighan CG, Easton J, Roberts S, Heatley SL, Ma J, Rusch MC, Chen
K, Harris CC, Ding L et al. CREST maps somatic structural variation in cancer
genomes with base-​pair resolution. Nat Methods 2011, 8(8):652–​654.
64. Layer RM, Chiang C, Quinlan AR, Hall IM. LUMPY: a probabilistic frame-
work for structural variant discovery. Genome Biol 2014, 15(6):R84.
65. Eisfeldt J, Vezzi F, Olason P, Nilsson D, Lindstrand A. TIDDIT, an efficient and
comprehensive structural variant caller for massive parallel sequencing data.
F1000Research 2017, 6:664.
66. pbsv (https://​git​hub.com/​Pac​ific​Bios​cien​ces/​pbsv)
67. Sedlazeck FJ, Rescheneder P, Smolka M, Fang H, Nattestad M, von Haeseler A,
Schatz MC. Accurate detection of complex structural variations using single-​
molecule sequencing. Nat Methods 2018, 15(6):461–​468.
68. Ebert P, Audano PA, Zhu Q, Rodriguez-​Martin B, Porubsky D, Bonder MJ,
Sulovari A, Ebler J, Zhou W, Serra Mari R et al. Haplotype-​resolved diverse
human genomes and integrated analysis of structural variation. Science 2021,
372(6537):eabf7117.
69. Gardner EJ, Lam VK, Harris DN, Chuang NT, Scott EC, Pittard WS, Mills
RE, Genomes Project C, Devine SE. The Mobile Element Locator Tool
(MELT): population-​scale mobile element discovery and biology. Genome Res
2017, 27(11):1916–​1929.
70. Tham CY, Tirado-​Magallanes R, Goh Y, Fullwood MJ, Koh BTH, Wang W, Ng
CH, Chng WJ, Thiery A, Tenen DG et al. NanoVar: accurate characterization of
patients’ genomic structural variants using low-​depth nanopore sequencing.
Genome Biol 2020, 21(1):56.
234 Next-Generation Sequencing Data Analysis

71. Cretu Stancu M, van Roosmalen MJ, Renkens I, Nieboer MM, Middelkamp
S, de Ligt J, Pregno G, Giachino D, Mandrile G, Espejo Valle-​ Inclan J
et al. Mapping and phasing of structural variation in patient genomes using
nanopore sequencing. Nat Commun 2017, 8(1):1326.
72. Zhou W, Emery SB, Flasch DA, Wang Y, Kwan KY, Kidd JM, Moran JV,
Mills RE. Identification and characterization of occult human-​specific LINE-​
1 insertions using long-​read sequencing technology. Nucleic Acids Res 2020,
48(3):1146–​1163.
73. Heller D, Vingron M. SVIM: structural variant identification using mapped
long reads. Bioinformatics 2019, 35(17):2907–​2915.
74. Gong L, Wong CH, Cheng WC, Tjong H, Menghi F, Ngan CY, Liu ET, Wei
CL. Picky comprehensively detects high-​ resolution structural variants in
nanopore long reads. Nat Methods 2018, 15(6):455–​460.
75. Cleal K, Baird DM. Dysgu: efficient structural variant calling using short or
long reads. Nucleic Acids Res 2022, 50(9):e53.
76. Zheng GX, Lau BT, Schnall-​ Levin M, Jarosz M, Bell JM, Hindson CM,
Kyriazopoulou-​Panagiotopoulou S, Masquelier DA, Merrill L, Terry JM et al.
Haplotyping germline and cancer genomes with high-​throughput linked-​read
sequencing. Nat Biotechnol 2016, 34(3):303–​311.
77. Zhang F, Christiansen L, Thomas J, Pokholok D, Jackson R, Morrell N, Zhao Y,
Wiley M, Welch E, Jaeger E et al. Haplotype phasing of whole human genomes
using bead-​based barcode partitioning in a single tube. Nat Biotechnol 2017,
35(9):852–​857.
78. Wang O, Chin R, Cheng X, Wu MKY, Mao Q, Tang J, Sun Y, Anderson E, Lam
HK, Chen D et al. Efficient and unique cobarcoding of second-​generation
sequencing reads from long DNA molecules enabling cost-​ effective and
accurate sequencing, haplotyping, and de novo assembly. Genome Res 2019,
29(5):798–​808.
79. Chen Z, Pham L, Wu TC, Mo G, Xia Y, Chang PL, Porter D, Phan T, Che H, Tran
H et al. Ultralow-​input single-​tube linked-​read library method enables short-​read
second-​generation sequencing systems to routinely generate highly accurate and
economical long-​range sequencing information. Genome Res 2020, 30(6):898–​909.
80. Long Ranger (https://​git​hub.com/​10XG​enom​ics/​lon​gran​ger)
81. Abyzov A, Urban AE, Snyder M, Gerstein M. CNVnator: an approach to dis-
cover, genotype, and characterize typical and atypical CNVs from family and
population genome sequencing. Genome Res 2011, 21(6):974–​984.
82. Xie C, Tammi MT. CNV-​seq, a new method to detect copy number variation
using high-​throughput sequencing. BMC bioinformatics 2009, 10:80.
83. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: Genome-​wide copy
number detection and visualization from targeted DNA sequencing. PLoS
Comput Biol 2016, 12(4):e1004873.
84. Klambauer G, Schwarzbauer K, Mayr A, Clevert DA, Mitterecker A,
Bodenhofer U, Hochreiter S. cn.MOPS: mixture of Poissons for discovering
copy number variations in next-​generation sequencing data with a low false
discovery rate. Nucleic Acids Res 2012, 40(9):e69.
85. Ivakhno S, Royce T, Cox AJ, Evers DJ, Cheetham RK, Tavare S. CNAseg-​-​a
novel framework for identification of copy number changes in cancer from
second-​generation sequencing data. Bioinformatics 2010, 26(24):3051–​3058.
Whole Genome/Exome Sequencing 235

86. Zhu M, Need AC, Han Y, Ge D, Maia JM, Zhu Q, Heinzen EL, Cirulli ET,
Pelak K, He M et al. Using ERDS to infer copy-​number variants in high-​
coverage genomes. Am J Hum Genet 2012, 91(3):408–​421.
87. Yoon S, Xuan Z, Makarov V, Ye K, Sebat J. Sensitive and accurate detection
of copy number variants using read depth of coverage. Genome Res 2009,
19(9):1586–​1592.
88. Boeva V, Popova T, Bleakley K, Chiche P, Cappo J, Schleiermacher G,
Janoueix-​Lerosey I, Delattre O, Barillot E. Control-​FREEC: a tool for assessing
copy number and allelic content using next-​generation sequencing data.
Bioinformatics 2012, 28(3):423–​425.
89. Alkan C, Kidd JM, Marques-​Bonet T, Aksay G, Antonacci F, Hormozdiari F,
Kitzman JO, Baker C, Malig M, Mutlu O et al. Personalized copy number and
segmental duplication maps using next-​generation sequencing. Nat Genet
2009, 41(10):1061–​1067.
90. Chiang DY, Getz G, Jaffe DB, O’Kelly MJ, Zhao X, Carter SL, Russ C,
Nusbaum C, Meyerson M, Lander ES. High-​resolution mapping of copy-​
number alterations with massively parallel sequencing. Nat Methods 2009,
6(1):99–​103.
91. Miller CA, Hampton O, Coarfa C, Milosavljevic A. ReadDepth: a parallel R
package for detecting copy number alterations from short sequencing reads.
PLoS One 2011, 6(1):e16327.
92. Roller E, Ivakhno S, Lee S, Royce T, Tanner S. Canvas: versatile and scalable
detection of copy number variants. Bioinformatics 2016, 32(15):2375–​2377.
93. Dharanipragada P, Vogeti S, Parekh N. iCopyDAV: Integrated platform for
copy number variations-​Detection, annotation and visualization. PLoS One
2018, 13(4):e0195334.
94. Cameron DL, Di Stefano L, Papenfuss AT. Comprehensive evaluation and
characterisation of short read general-​purpose structural variant calling soft-
ware. Nat Commun 2019, 10(1):3240.
95. Kosugi S, Momozawa Y, Liu X, Terao C, Kubo M, Kamatani Y. Comprehensive
evaluation of structural variation detection algorithms for whole genome
sequencing. Genome Biol 2019, 20(1):117.
96. Wong K, Keane TM, Stalker J, Adams DJ. Enhanced structural variant and
breakpoint detection using SVMerge by integration of multiple detection
methods and local assembly. Genome Biol 2010, 11(12):R128.
97. Zarate S, Carroll A, Mahmoud M, Krasheninina O, Jun G, Salerno WJ, Schatz
MC, Boerwinkle E, Gibbs RA, Sedlazeck FJ. Parliament2: Accurate structural
variant calling at scale. GigaScience 2020, 9(12):giaa145.
98. Becker T, Lee WP, Leone J, Zhu Q, Zhang C, Liu S, Sargent J, Shanker K, Mil-​
Homens A, Cerveira E et al. FusorSV: an algorithm for optimally combining data
from multiple structural variation detection methods. Genome Biol 2018, 19(1):38.
99. Jeffares DC, Jolly C, Hoti M, Speed D, Shaw L, Rallis C, Balloux F, Dessimoz
C, Bahler J, Sedlazeck FJ. Transient structural variations have strong effects
on quantitative traits and reproductive isolation in fission yeast. Nat Commun
2017, 8:14061.
100. Mohiyuddin M, Mu JC, Li J, Bani Asadi N, Gerstein MB, Abyzov A, Wong
WH, Lam HY. MetaSV: an accurate and integrative structural-​variant caller
for next generation sequencing. Bioinformatics 2015, 31(16):2741–​2744.
236 Next-Generation Sequencing Data Analysis

101. Medvedev P, Fiume M, Dzamba M, Smith T, Brudno M. Detecting copy


number variation with mated short reads. Genome Res 2010, 20(11):1613–​1622.
102. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of gen-
etic variants from high-​throughput sequencing data. Nucleic Acids Res 2010,
38(16):e164.
103. Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, Land SJ, Lu
X, Ruden DM. A program for annotating and predicting the effects of single
nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila
melanogaster strain w1118; iso-​2; iso-​3. Fly 2012, 6(2):80–​92.
104. McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A, Flicek P,
Cunningham F. The Ensembl Variant Effect Predictor. Genome Biol 2016,
17(1):122.
105. Hinrichs AS, Raney BJ, Speir ML, Rhead B, Casper J, Karolchik D, Kuhn
RM, Rosenbloom KR, Zweig AS, Haussler D et al. UCSC Data Integrator and
Variant Annotation Integrator. Bioinformatics 2016, 32(9):1430–​1432.
106. SeattleSeq (http://​snp.gs.was​hing​ton.edu/​Seat​tleS​eqAn​nota​tion​138/​)
11 
Clinical Sequencing and Detection
of Actionable Variants

Next-​ generation sequencing has not only altered the landscape of life
science research, its impact on clinical diagnosis, prognosis, and interven-
tion selection has also become increasingly evident. The launch of precision
or personalized health initiatives worldwide is a testament to the power of
NGS in improving human health, and also a key driver for integrating NGS
into medical practice. From the rapid development of clinical sequencing, it
is apparent that personal genome information guided medicine is the future
of medical practice. Compared to research-​oriented NGS, clinical sequencing
is subjected to more regulations as required for other clinical tests of patient
samples to ensure accurate and reliable results. In the United States, clin-
ical sequencing is mostly regulated by the Food and Drug Administration
(FDA) and Centers for Medicare & Medicaid Services through the Clinical
Laboratory Improvement Amendments (CLIA). Many countries around the
world and international organizations such as ISO have similar regulations.
Diagnosis, prognosis, and treatment of oncologic and pediatric diseases are
two exemplary areas that have seen great benefits from clinical sequencing.
As cancer is a disease of the genome, NGS is well suited to unravel tumor
heterogeneity and classify tumors into different types or subtypes based on
what genomic variants they possess [1]. Sequencing of various oncological
gene panels, whole exome, and increasingly whole genome has become
more and more commonplace, and provided much needed guidance for clin-
ical actions. Tumor mutation burden, an overall index of the total amount
of nonsynonymous mutations in a genome measured by NGS, serves as a
good indicator of immunotherapy efficacy [2]. For pediatric patients, espe-
cially those in the neonatal intensive care unit (NICU), speedy diagnosis and
treatment are essential, which require rapid sequencing and data processing.
The use of NGS in the NICU setting is a test to its speed, accuracy, and overall
utility in meeting clinical needs. The development of rapid genome sequen-
cing pipelines, including bioinformatics, has been proven to decrease infant
morbidity and at the same time lead to cost savings [3].
Often different from a research setting, the immediate goal of clinical sequen-
cing is to identify disease-​causing variant(s) based on which a treatment plan
can be decided. The major challenge to achieve this goal is how to identify the
causal variant(s) for the primary indication from thousands or even millions

DOI: 10.1201/9780429329180-14 237


238 Next-Generation Sequencing Data Analysis

of called variants. As focused on in this chapter, this requires a multi-​faceted


approach and corresponding tools to filter, rank, and prioritize the variants.
This chapter starts with patient sample collection and sequencing approaches.
After the steps on how to find actionable variants are detailed, the current
variant classification system based on their pathogenicity is presented. This
is followed by conduct of clinical review and generation of clinical reports.
Validation of a bioinformatics pipeline for clinical sequencing is presented at
the end.

11.1 Clinical Sequencing Data Generation


11.1.1 Patient Sample Collection
For the diagnosis of Mendelian disorders, i.e., those caused by inherited
mutation(s) in a single gene, various types of patient samples may be used.
Among the most widely used are peripheral blood (including dried blood
spots), saliva, buccal swab, etc. For diseases that require identification of som-
atic mutations, such as solid tumors, tissue biopsies collected for pathological
examinations, such as hematoxylin & eosin staining, immunohistochemistry,
or fluorescent in situ hybridization, can be used. To detect somatic mutations,
besides DNA extracted from such pathogenic tissues (or cells), matched
normal control DNA is also needed. Such control DNA is often prepared
from peripheral blood for most diseases, including solid tumors. For hema-
tologic malignancies, however, control DNA may come from buccal swab,
saliva, nail, hair follicle, skin biopsy, etc. In cases where normal control DNA
is not available, the use of DNA from pathogenic tissues/​cells alone can
lead to overestimation of somatic mutations [4], and/​or missing of germline
mutations that may underlie genetic predisposition to other conditions.
Among the most abundant and readily available tissue biopsies are those
that are chemically fixed with traditional fixatives such as formaldehyde.
Such fixatives, while preserving tissue morphology for pathological exam-
ination, cause DNA damage ranging from chemical modifications (such as
formation of adducts, intra-​and inter-​strand cross-​links, as well as crosslinks
with proteins), fragmentation, and strand separation [5]. This damage may
introduce up to 30% artifactual mutation profiles that were not found in the
original tissue [6]. Therefore, caution must be used when starting with such
previously fixed archival tissues in order to minimize sequence artifacts [7].
Newer, alternative fixatives that have been shown to preserve DNA quality,
protein antigenicity, and at the same time morphology [5] should be used
for new collections. If archival formalin-​ fixed paraffin-​
embedded (FFPE)
tissues need to be used, strategies developed to obtain the best NGS sample
quality possible, including deparaffinization and FFPE-​compatible nucleic
Clinical Sequencing and Detection of Actionable Variants 239

acid extraction [8], can help decrease the frequency of such false-​positive
discoveries.
Other surgical biopsy materials, including fine needle aspiration and core
needle biopsy, are also regularly used to sample discrete site(s) of diseased
tissue. A potential pitfall here is that they may not be representative of the
remaining, unsampled parts of the tissue. Some diseases, with cancer being
the best known, are characterized by clonality and cellular heterogeneity.
For these diseases, the sampling with tissue biopsies may not fully reveal
the entire set of mutations. Also because of its invasiveness, obtaining tissue
biopsies is not suitable for sample collection at regular intervals for the pur-
pose of tracking disease progression and monitoring treatment outcome.
Liquid biopsy represents a more recently developed sample type that is
less invasive and therefore better suited for real-​time, longitudinal clinical
tracking and monitoring. Instead of sampling diseased tissue directly, liquid
biopsy collects cells or DNA that are shed from diseased tissue into the
blood or other bodily fluid (such as urine). For example, circulating tumor
cells (CTCs), or circulating cell-​free DNA (cfDNA), are increasingly used as
input. Because the amount of cfDNA in the plasma or other bodily fluid
is rather low, of which circulating tumor DNA (ctDNA) only constitutes a
minor fraction (<0.1–​10%), it is crucial to use a cfDNA extraction method
that provides high efficiency and recovery [9, 10]. While cancer patients usu-
ally have more cfDNA than healthy individuals [11], the extraction yield
is generally within the range from below 10 ng to 100 ng cfDNA per mL
plasma (usually below 20 ng). The fragment size of cfDNA is mostly within
the range of 160–​200 bp [12]. Side-​by-​side comparisons have demonstrated
high levels of concordance on the detection of mutations between cfDNA
and matched tissue biopsies [11, 13]. Because of its low invasiveness and
high accuracy, liquid biopsy has been used for multiple clinical applications,
including detection of minimal residual disease for patients that are in remis-
sion, or early screening of healthy individuals before a disease manifests
itself [14].
Prior to sample collection, patients need to counseled and informed con-
sent must be obtained. Besides the affected individual (proband), often
the proband’s parents and/​or other family members may also need to be
sequenced. This is especially required to determine whether a mutation is
passed on from parents or formed de novo, for which samples are collected
from the proband and their biological parents for trio sequencing. During
counseling, the purpose of performing genetic testing and the types of results
anticipated are conveyed to the patient and their family members. In addition,
the patient and family members are also informed of the test’s limitations and
potential risks. For example, the test may not reveal a genetic link, the inter-
pretation may involve uncertainty, or the findings may be distressing instead
of reassuring. Further, interpretation of the results may change over time,
and the test results may have implications to other untested family members
and their lives.
240 Next-Generation Sequencing Data Analysis

Post sample collection, to avoid potential sample mix-​up, patient sample


transfer, storage, and sequencing library preparation (detailed below) should
be systematically tracked. This tracking can be achieved through performing
routine sample genotyping using microarray or STR (Short Tandem Repeats)
marker profiling, and subsequently comparing the genotyping result to that
directly generated from the sequencing data. When proband-​parent trios as
well as other family members are sequenced, parentage and relatedness need
to be checked using standard kinship determination methods [15].

11.1.2 Library Preparation and Sequencing Approaches


What type of NGS library to make from the DNA extracted from collected
patient samples, and how to make them to best inform clinical decision
making, require consideration of multiple factors. These factors include
patient sample type, quality and quantity of the extracted DNA, genomic
coverage needed, type(s) of variant to be detected, and limit of detection
(LOD) to be achieved. For example, if liquid biopsy is used as input, only
limited quantity of target DNA is available, which requires a sequencing
library preparation procedure with high sensitivity. If archival FFPE tissue
is used, the quality of the extracted DNA might be low and needs to be
evaluated first using fragment analysis and/​or quantitative PCR. If low-​
quality FFPE-​derived DNA sample has to be used, more input DNA and/​or
increased PCR cycle number are usually needed for library prep. To mitigate
the effects of fixative-​caused DNA damage, DNA repair may be conducted
prior to library making with the use of uracil-​DNA glycosylase, or a mixture
of multiple DNA repair enzymes.
Depending on the quantity of starting DNA, the library preparatory pro-
cedure may involve PCR amplification or be PCR-​free. A PCR-​free procedure
can avoid amplification-​caused artifacts, including PCR stochasticity, poly-
merase errors, and amplification biases (such as different amplification effi-
ciencies due to variation in fragment length and GC content), but does require
larger amounts of DNA to start, e.g., 1–​2 µg. For lower input quantity, e.g., at
the ng or even pg level, PCR amplification is needed to boost library yield.
With continuous advancements in NGS library construction technology, gen-
erating enough library molecules from ultra-​low amounts of DNA, such as
those extracted from very limited patient biopsy materials or liquid biopsy,
is no longer a major challenge. With commercial library prep kits currently
available (as of 2022), libraries can be prepared from as little as 10 pg of DNA.
With whole genome amplification using approaches such as multiple dis-
placement amplification, libraries can even be prepared from a single cell that
has only two copies of genomic DNA (~6–​7 pg) [16].
Depending on genomic coverage needed, sequencing libraries for whole
genome, exome, or selected target genes (gene panel) may be prepared.
Whole genome sequencing enables generation of the most comprehensive
Clinical Sequencing and Detection of Actionable Variants 241

set of patient genetic variant information, encompassing coding and non-​


coding single nucleotide variants, indels, copy number variants, and struc-
tural variants. Exome sequencing, in comparison, focuses on detecting SNVs
and small indels in coding regions, while it also allows detection of CNVs
and SVs although at reduced power. With the continuous drop in sequen-
cing cost, there is an ongoing debate on the use of whole genome vs. exome
sequencing for patient sample sequencing. While WGS generates the most
comprehensive variant information for a patient, it requires significantly
more data storage and processing capabilities, and a significant number of
identified variants (especially those located in non-coding regions) is cur-
rently uninterpretable. Exome sequencing needs significantly less sequen-
cing power and informatics resources, generates more interpretable SNV
and indel information, but this comes at the cost of much reduced power
for CNV and SV detection. Targeted gene panel sequencing uses a different
approach interrogating only genes that are known to be associated with a
disease of interest. By focusing on selected genes, the amount of sequen-
cing required is further reduced and, as a result, high sequencing depth can
be readily accomplished thereby leading to increased detection sensitivity
and specificity. This is particularly helpful for making heterozygous calls,
detecting somatic mutations at low frequency, and analyzing heteroplasmic
mitochondrial DNA variants for diseases linked to dysfunctional mitochon-
drial metabolism. Because only disease-​associated genes are analyzed, the
demand on bioinformatics power is further reduced, and the result is more
likely to be interpretable. Among the widely available disease-​specific gene
panels are those built for precision oncology, including the Memorial Sloan
Kettering–​Integrated Mutation Profiling of Actionable Cancer Targets (MSK-​
IMPACT) and the FoundationOne CDx (F1CDx) panels, both of which are
approved by the FDA. These panels use hybridization probes to capture their
gene targets and are designed to measure SNVs, indels, CNVs, and SVs in
the genes they target, as well as microsatellite instability (MSI) and tumor
mutation burden (TMB).
Targeted gene panel sequencing is particularly suitable for detecting
low-​frequency variant alleles for demanding applications such as cancer
screening, minimal residual disease assessment, prenatal diagnostics, and
infectious agent detection. To reach the low levels of LOD needed in these
applications (e.g., variant allele frequency or VAF <5%), sequencing needs to
be performed at greater depth than that needed to detect germline mutations,
or somatic mutations from tissues enriched with pathogenic cells. For
example, based on calculation using the binomial distribution, a sequencing
depth of 1,650× is needed to detect mutations at ≥3% frequency with at least
30 mutation-​supporting reads [17]. In comparison, for detection of germline
mutations using WGS, the typical sequencing depth is 30×. To detect som-
atic mutations from tissues that contain at least 20% tumor cells using WGS,
a depth of 100× is needed along with at least 10 variant-​supporting reads,
242 Next-Generation Sequencing Data Analysis

with the matched control DNA sequenced to ≥30× [18]. Sequencing depth is
largely determined by the accuracy of sequencing (e.g., 0.1–​1% of error rate on
Illumina sequencers, refer to Chapter 4 for more details), and other molecular
steps such as PCR amplification during library preparation. To reach even
lower LOD (e.g., VAF <1%), strategies to improve NGS accuracy and reduce
PCR errors have been deployed, which include employment of signal-​to-​
noise correction methodologies and single-​molecule consensus sequencing
schemes [19]. One of such strategies is called Duplex Sequencing built on
the use of molecular barcoding. Molecular barcoding utilizes the so-​called
unique molecular identifiers (or UMIs) to label single molecules prior to PCR
amplification. From sequences generated from PCR duplicates that carry the
same UMI, i.e., those derived from the same molecule, consensus sequence
is reached that corrects errors introduced during amplification (except the
first cycle) as well as random sequencing errors. In Duplex Sequencing, the
two strands of the original DNA duplex generate two separate consensus
sequences, and comparing them leads to generation of duplex consensus
sequence, further removing errors introduced during the first PCR cycle.
With Duplex Sequencing, somatic mutations that occur at a frequency of 10-​5
or lower can be detected with high confidence [20]. Because it relies on single
molecule consensus sequence generation through sequencing multiple amp-
lified copies of the same original molecule in a strand-​specific fashion and
then later collapsing them, this technique requires a lot more reads than con-
ventional NGS to achieve lower LOD levels.
For QC/​QA during library preparation and sequencing, sample and
data quality needs to be checked at multiple steps. Prior to proceeding to
library preparation, input DNA quality and quantity need to be assessed
against pre-​defined criteria, which may vary depending on the intended
detection targets of the sequencing assay. For example, for detection of
structural variants, which needs long-​range genomic information, FFPE
samples are not recommended even with the use of remedial measures
mentioned above. Once an appropriate library preparatory workflow is
chosen, it needs to be standardized to ensure consistent performance,
and multiple QC/​QA steps should be specified to monitor results of the
workflow at key junctures, eventually library yield and fragment size
range. To help assess workflow performance, reference DNA samples with
known variants can be used as positive control. Such reference samples
include well-​characterized reference DNA from the Genome in a Bottle
(GIAB) consortium supported by the U.S. National Institute of Standards
and Technology [21], engineered DNA that contains clinically relevant
synthetic variants at pre-​defined allele frequencies (available from com-
mercial sources), and clinical samples that have been analyzed by another
CLIA-​accredited NGS lab. Sequencing run quality metrics, such as the per-
centage of reads over Q30 and overall error rate, need to be monitored and
must pass a pre-​defined quality threshold.
Clinical Sequencing and Detection of Actionable Variants 243

11.2 Read Mapping and Variant Calling


Identification of disease-​causing variants from sequencing data is a multi-​step
process. Chapters 5 and 10 cover many of the upstream steps in this process,
including read mapping and variant calling. As these steps also apply to clin-
ical samples, they are not to be repeated here. In the clinical setting, how-
ever, speed and turnaround time are often of essence for clinicians to make
diagnosis, prognosis, and treatment decisions. Various hardware or algo-
rithmic implementation strategies have been devised to speed up the read
mapping and variant calling process. These strategies include: (1) deployment
of specialized hardware, such as field programmable gate arrays (FPGAs), as
used by the DRAGEN (Dynamic Read Analysis for GENomics) platform for
significantly accelerated mapping and variant calling [22]; (2) optimization
for speed of highly efficient workflows such as Genalice [23] and Sentieon
[24]; (3) real-​
time [25] or pulsed [26] read mapping and variant calling
while sequencing is still underway; or (4) employment of a sequencing tech-
nology that has concurrent analysis capabilities while sequencing, like Oxford
Nanopore Technology. With these strategies, it has become a reality to receive
provisional diagnosis and prognosis from clinical sample collection within
20 hours [27]. After mapping of reads to the reference genome, median (or
mean) depth of coverage, uniformity of coverage, and on-​target capture rate
(for exome and gene panel sequencing) need to be examined and compared to
pre-​defined QC threshold. Next the called variants are subjected to an exten-
sive process of filtering and annotation to produce a short list of actionable
variants, which as detailed next is the major focus of this chapter.

11.3 Variant Filtering
The procedure from a long list of called variants to a clinical testing report
(Figure 11.1) is the major focus of this chapter. Among the large number
of called variants, most are benign and do not have an impact on human
health. For example, a typical WGS of an individual’s germline DNA usually
identifies 5 million or more variants, of which only 30,000 or more are in
protein-​coding regions. Of these variants, ~10,000 represent missense amino
acid substitutions, aberrant splicing sites, or small indels [28]. These variants
are further narrowed down to produce a short list of clinically relevant and
actionable variants, to assist clinicians in disease diagnosis, prognosis, and
treatment. This multi-​step process requires a multitude of tools and databases
to screen the called variants based on their frequency, functional consequence,
known linkage to human disease(s), and match of clinical phenotype, as well
244 Next-Generation Sequencing Data Analysis

Called Variants

Variant Filtering
- Frequency
- Functional Impact
- Known Evidence
- Phenotype Match
- Inheritance Mode

Variant Ranking
& Prioritization

Variant Pathogenicity
Classification

Expert Review

Testing Report Generation

Variant Validation

Periodic Report Updates


and Patient Reconact

FIGURE 11.1
Clinical sequencing general data analytic workflow starting from variant calls.

as mode of inheritance. Besides these in silico variant filtering steps, wet lab
strategies such as pedigree sequencing can also help to reliably and signifi-
cantly reduce the number of potential candidate variants.
Presented below are major steps of the variant filtering workflow. Prior to
performing these steps, preliminary variant screening is needed to filter out
variants that do not pass a predefined variant call quality threshold. While
the following filtering steps are usually performed on all variants that pass
the threshold, these steps can also be applied to a pre-​selected list of genes
that are known to be associated with the phenotype/​disease of the patient,
for the purpose of minimizing incidental findings and reducing analytic
burden. Each of the filtering steps detailed below focuses on one relevant
aspect of a variant to its potential role in underlying the disease or phenotype
Clinical Sequencing and Detection of Actionable Variants 245

observed in the patient. In combination, the multi-​faceted filtering provided


by these steps will become the basis for generating a list of top candidates for
additional vetting toward clinical action.

11.3.1 Frequency of Occurrence
Many called variants are too common to be consistent with the low incidence
of a genetic disease. To identify disease-​causing variant(s), such common
variants need to be filtered out. While the threshold for occurrence frequency
can be set at different levels, an MAF (minor allele frequency) of less than 1%
is often used. To determine the occurrence frequency of a called variant in
the general population, large databases of human genetic variations, such as
gnomAD [29], the 1000 Genomes Project (1KGP) database [30], TOPMed [31],
UK10K [32], and NHLBI Exome Sequencing Project (ESP) [33], are often used
(Table 11.1, Page 249). These databases contain mostly SNVs and short indels.
Some of these databases, such as gnomAD and 1KGP, also contain common
structural variants, but most SVs are catalogued by specific databases including
the Database of Genomic Variants (DGV) [34], Database of Chromosomal
Imbalance and Phenotype in Humans using Ensembl Resources (DECIPHER)
[35], and dbVar [36]. It should be noted that variant allele frequency is often
population specific. For example, the minor allele of an SNV in the EML6 gene,
rs17046386 (A>G), is not rare in African populations, but rare in non-​African
populations. Therefore, the ancestral background of the affected individual
should be taken into consideration in this step.

11.3.2 Functional Consequence
Variants that change amino acid residues in the active site of a protein may
significantly affect its function. Variants located at other conservative base
positions, such as those that affect gene transcript splicing or gene transcrip-
tion initiation, may also exert significant effects on gene product. On the other
hand, functional significance of variants that fall into intergenic regions are
often hard to assess. To sort variants based on their genomic locations, e.g.,
those in protein-​coding, regulatory (e.g., intron, splicing site, 5’ or 3’ UTR,
promoter, etc.), or intergenic non-​coding regions, variant annotation tools
such as ANNOVAR, VEP, or VariantAnnotation [37] can be used. Intergenic
or non-​coding variants are usually filtered out, unless they are predicted to
have regulatory functions such as affecting gene splicing. To predict potential
variant pathogenicity caused by altered splicing, SpliceAI [38], MaxEntScan
[39], and NNSplice [40] are among the best performing methods, based on
currently available benchmarking studies [41–​44]. For amino acid-​altering
variants, a variety of tools are available to predict their potential impacts to
help determine whether they should be filtered out. Such tools, based on their
underlying algorithm, can be divided into three groups: function prediction,
246 Next-Generation Sequencing Data Analysis

evolutionary conservation, and ensemble. Function prediction methods, typ-


ically based on biochemical properties of amino acids, include PolyPhen [45],
PROVEAN [46], SIFT [47], MutationTaster [48], MutationAssessor [49], VEST
[50], FATHMM [51], MutPred [52], and LRT [53]. Methods that are based
on evolutionary sequence conservation include phyloP [54], phastCons
[54], GERP [55], and SiPhy [56]. Those that employ the ensemble approach
aggregate and integrate information about a variant from different sources
for variant prioritization to help identify disease-​causing genes. Examples
of these methods are CADD [57], M-​CAP [58], MetaRNN [59], REVEL [60],
Eigen [61], and VAAST [62].

11.3.3 Existing Evidence of Relationship to Human Disease


There are multiple community efforts to catalog the relationship of human
diseases to individual genes and the variants they harbor. For example,
ClinVar [63], Human Gene Mutation Database (HGMD) [64], and Online
Mendelian Inheritance in Man (OMIM) [65] are databases that aggregate
genes/​variants and their relationship to human health. Besides these com-
prehensive databases, there are also efforts to collect variants as they relate to
particular classes of diseases. For example, COSMIC (Catalogue Of Somatic
Mutations In Cancer) [66], National Cancer Institute’s GDC (Genomic Data
Commons) [67], and American Association for Cancer Research (AACR)’s
GENIE (Genomics Evidence Neoplasia Information Exchange) [68], catalog
variants that appear in various cancers. Among variants identified in a
patient, if certain variant(s) have already been cataloged by a relevant data-
base, the chance of the variant(s) bearing a relationship to the patient’s dis-
ease becomes higher. Most of these databases can be queried through web
interface providing user-​friendliness and most up-​to-​date information. For
efficiency and consistency, the variant-​disease relationship contained in these
databases can also be accessed through the use of Application Programming
Interface (API), or downloaded for local deployment.

11.3.4 Clinical Phenotype Match


Identification of actionable variants starts even before sequencing, as a
patient’s clinical phenotype can greatly aid variant filtering. To standardize
clinical phenotyping, the Human Phenotype Ontology (HPO) project provides
structured vocabulary to describe phenotypic abnormalities manifested in
human diseases, thereby enabling integrated analysis of semantic pheno-
typic information with NGS genotypic data. Once appropriate HPO terms are
identified to capture the clinical phenotype of a patient, a number of tools can
then be used to filter variants based on such HPO terms. These tools include
Exomiser [69], eXtasy [70], Genomiser [71], Phenolyzer [53], Phenomizer [72],
PhenIX [28], Phevor [73], Phen-​Gen [74], VarElect [75], and DeepPVP [76].
Clinical Sequencing and Detection of Actionable Variants 247

These tools draw on current knowledge of the relationships between dis-


ease phenotypes and genetic variants to rank variants from a patient. Besides
using highly specific phenotypic terms, sometimes it also helps to expand
the search to include synonyms or more general terms at higher levels up
the Disease Ontology (DO) hierarchy. While the phenotype –​genetic variant
relationship is typically extracted from human disease databases such as
OMIM, Orphanet, DECIPHER, and ClinVar, they can also be extracted from
databases of model organisms, such as MGI (Mouse Genome Informatics) and
ZFIN (Zebrafish Information Network). Phenotypic information extracted
from these databases can be translated to human diseases using tools such as
PhenomeNET [77] and PHIVE (for mouse) or hiPHIVE (human/​interaction
PHIVE, for both mouse and zebrafish), both as parts of Exomiser [69].

11.3.5 Mode of Inheritance
Family medical history, if available, can also greatly aid the variant filtering
process. For a Mendelian disorder, the five basic modes of inheritance are
autosomal dominant, autosomal recessive, sex-​linked dominant, sex-​linked
recessive, and mitochondrial. Traditional pedigree analysis can lead to
revealing of the mode of inheritance of such a disorder. From variants called
from a proband and their pedigree (most commonly trio sequencing), those
that do not conform to the inheritance pattern are filtered out.

11.4 Variant Ranking and Prioritization


Using the multiple filters above, to eventually get down to a short list of clin-
ically actionable variants for reporting, filtered variants need to be ranked
and prioritized. To speed up this process, systems are needed to integrate the
large number of tools and databases above. Publicly available open-​source
systems, as well as commercially developed proprietary systems, are avail-
able to meet this need. Examples of publicly available tools include VarFish
[83], KGGSeq [84], MutationTaster2021 [48], VAAST Variant Prioritizer (VVP)
[85], VaRank [86], Variant Ranker [87], and Variant Prioritization Ordering
Tool (VPOT) [88]. Using as input VCF files and often patient’s clinical pheno-
type, these tools offer the capabilities to annotate, filter, rank, and prioritize
the imported variants. Many of the tools offer highly interactive filtering
affording users the flexibility to use different permutations of the various
filters on allele frequency, functional consequence, phenotypic association,
existing disease connection, and inheritance pattern. Some of these tools,
such as KGGSeq, MutationTaster2021, and Variant Ranker, also allow users
to identify variants in genes associated with a particular biological pathway,
248 Next-Generation Sequencing Data Analysis

Gene Ontology term, gene network, or displaying a particular expression pro-


file such as tissue specificity. To facilitate variants ranking, some of the tools,
exemplified by VaRank, Variant Ranker, and VPOT, generate an aggregated
score from the various scores computed by many of the tools summarized
on Table 11.1. Each of these scores provides information on one aspect of a
variant’s potential contribution to disease development, such as functional
consequence (e.g., from PolyPhen), sequence conservation (e.g., phyloP),
splicing site alteration (e.g., MaxEntScan), etc. Since the contribution of each
of these aspects to disease outcome may not be equal, different weights can
be applied to the different scores in order to produce a single aggregated
score to serve as an index of the variant’s overall pathogenicity. VPOT, as an
example, allows application of user defined weighting factors to each score
to calculate a final, aggregated score for variant ranking and prioritization.

11.5 Classification of Variants Based on Pathogenicity


11.5.1 Classification of Germline Variants
In terms of the potential role they play in disease development, ranked
variants can be anywhere in a continuous range from benign to patho-
genic. To proceed to clinical reporting, they need to be classified based on
existing evidence of their pathogenicity. In the United States, using the
current joint guidelines from the American College of Medical Genetics
and Genomics (ACMG) and the Association for Molecular Pathology
(AMP), germline variants for Mendelian diseases are classified using
a five-​tier system. This system places variants into one of the following
tiers: (1) Pathogenic: directly contributing to the development of the dis-
ease; (2) Likely Pathogenic: highly possible (with over 90% certainty) to
cause the disease; (3) Uncertain Significance: currently available data and
literature being inconclusive to declare the pathogenicity of the variant;
(4) Likely Benign: high possible (with over 90% certainty) that the variant
is not the cause of the disease; and (5) Benign: not disease causing.
To standardize classification of variants into these five tiers, specific
evidence-​based criteria have been established. For pathogenic variants, the
evidence of pathogenicity is divided into four levels: (1) Very Strong (coded
as PVS1) –​these are null variants (nonsense, frameshift, canonical +​/​–​1 or 2
splicing sites, initiation codon, mono-​or multi-​exon deletion, etc.) in a gene
of which loss of function is established as a known mechanism of disease;
(2) Strong (PS) –​ranges from missense variants causing the same amino
acid alteration as a pathogenic variant established earlier, to variants whose
prevalence level is significantly higher in affected individuals than controls;
(3) Moderate (PM) –​encompasses variants from those located in a gene’s hot
newgenrtpdf
TABLE 11.1

Clinical Sequencing and Detection of Actionable Variants


Major Tools and Databases for Filtering Variants Using Various Criteria
Name Full Name Description Reference

Variant Allele Frequency Databases


1KGP 1000 Genomes Project Contains genomes of thousands of individuals from dozens of [30]
populations, reconstructed with low-​coverage WGS, deep WES, and
dense array genotyping
gnomAD Genome Aggregation Database Aggregation of harmonized exome and genome sequencing data from a [29]
variety of large-​scale sequencing projects
UK10K UK10,000 Genomes/​Exomes Project Includes WGS data from ~4,000 healthy phenotyped individuals, and [32]
WES data from ~6,000 individuals with rare disease, severe obesity, and
neurodevelopmental disorders
TOPMed Trans-​Omics for Precision Medicine WGS and other -​omics data collected for heart, lung, blood, and sleep [31]
(Supported by NIH/​NHLBI) disorders from large and diverse cohorts, data available via dbGaP
ESP Exome Sequencing Project WES data collected to discover variants associated with heart, lung, and [33]
(Supported by NIH/​NHLBI) blood diseases or traits, available via dbGaP
Predicted Functional Consequence (Missense Variants)
PolyPhen Polymorphism Phenotyping Predicts possible effect of amino acid substitutions on protein function [45]
based on structural attributes and conservation profiles. Scores of 0.0–​
0.15 are thought as benign while 0.15–​1.0 as damaging
SIFT Sorting Intolerant From Tolerant Classifies an amino acid coding change as tolerated or deleterious [47]
based on protein sequence conservation captured into a SIFT score (0–​
0.05: deleterious; 0.05–​1.0: tolerated)
PROVEAN Protein Variation Effect Analyzer Computes a pairwise sequence alignment score to quantify the impact of [46]
a variant on protein function, with low score representing deleterious
and high score neutral effect
(continued)

249
newgenrtpdf
TABLE 11.1 (Continued)

250
Major Tools and Databases for Filtering Variants Using Various Criteria
Name Full Name Description Reference

MutationTaster NA Deploys machine learning to predict whether a coding or non-​coding [48]


variant is deleterious, in part based on evolutionary conservation,
variant frequency, and patient phenotype
MutationAssessor NA Derives functional impact score for amino acid residue changes, based on [49]
sequence conservation at the residue positions determined by aligning
families and subfamilies of sequence homologs within and between
species
VEST Variant Effect Scoring Tool Applies a Random Forest classifier to 86 variant features to generate [50]
a pathogenicity score and an associated p-​value to determine the
confidence of calling a variant pathogenic

Next-Generation Sequencing Data Analysis


FATHMM Functional Analysis Through Predicts functional effects of missense variants by integrating protein [51]
Hidden Markov Models sequence conservation with pathogenicity weights, which represent the
general tolerance to mutations of proteins and their domains
LRT Likelihood Ratio Test Identifies deleterious mutations using the likelihood ratio test, based on [53]
the null model that each codon evolves neutrally with no difference in
the rate of missense to synonymous substitution
MutPred NA Quantifies pathogenicity of amino acid substitutions, and models their [52]
impacts on protein structure and function
Evolutionary Conservation
phyloP Phylogenetic P-​value Measures evolutionary conservation at an aligned position against a [54]
model of neutral drift. Positive p-​values indicate conservation while
negative p-​values fast evolution
phastCons NA Identifies conserved sequences through fitting a phylogenetic hidden [54]
Markov model to multiple sequence alignment data
GERP Genomic Evolutionary Rate Uses maximum likelihood evolutionary rate estimation to identify [78]
Profiling evolutionarily constrained sequence elements through multiple
alignments
Clinical Sequencing and Detection of Actionable Variants
SiPhy SIte-​specific Predicts constrained sequence elements using a hidden Markov model [56]
PHYlogenetic analysis based on the pattern of base substitutions from sequence alignments
Ensemble Methods
CADD Combined Annotation Dependent Integrates diverse annotations about a variant, including evolutionary [57]
Depletion constraint, functional prediction, epigenetic marking, etc., into a single
score to measure deleteriousness
M-​CAP Mendelian Clinically Applicable Uses a gradient boosting tree classifier to separate pathogenic and benign [58]
Pathogenicity variants using both existing pathogenicity scores (e.g., CADD, SIFT, and
PolyPhen) and direct measures of sequence conservation
MetaRNN NA Employs a recurrent neural network model to integrate major existing [59]
variant annotation scores, sequence conservation, and allele frequency
to generate a meta-​score for variant pathogenicity
REVEL Rare Exome Variant Ensemble Uses Random Forest to predict pathogenicity of missense variants based [60]
Learner on features from an ensemble of existing methods
Eigen NA Estimates predictive accuracy of various existing functional annotations [61]
of a variant, and uses such estimates to derive an aggregate functional
score
VAAST Variant Annotation, Analysis and Incorporates information on amino acid substitution severity, variant [62]
Search Tool frequency, and phylogenetic conservation for variant prioritization
Predicted Splicing Alterations
SpliceAI NA Employs deep neural network to predict variants that cause cryptic [38]
splicing, through modeling of long-​range genomic sequence information
around the variant site
MaxEntScan Maximum Entropy Scan Generates splice prediction scores through modeling of short sequence [39]
motifs that accounts for dependencies between non-​adjacent as well as
adjacent positions, based on maximum entropy distribution
NNSplice Splice Site Prediction by Neural Uses neural network to predict splice site through exploiting pairwise [40]
Network correlations between adjacent nucleotides
(continued)

251
newgenrtpdf
TABLE 11.1 (Continued)

252
Major Tools and Databases for Filtering Variants Using Various Criteria
Name Full Name Description Reference

Databases of Variants with Known Disease Associations


ClinVar NA Public database maintained by NCBI that contains both germline and [79]
somatic variants in the nuclear and mitochondrial genomes, and their
relationships to human diseases and phenotypes
COSMIC Catalogue Of Somatic Mutations In Expert curated database on the effects of somatic mutations across human [66]
Cancer cancers
OMIM Online Mendelian Inheritance in Compendium of human genes and genetic disorders, with curated [65]
Man descriptions of genes, phenotypes, and their relationships in a
structured free-​text format
HGMD Human Gene Mutation Database Collates published germline mutations in nuclear genes and their [64]

Next-Generation Sequencing Data Analysis


relationships to inherited human diseases
DGV Database of Genomic Variants Provides a curated catalog of genomic structural variants in health [34]
controls
DECIPHER Database of Chromosomal Repository of CNVs and other genomic variants associated with disease [80]
Imbalance and Phenotype in phenotypes, to aid in finding pathogenic variants in patients of rare
Humans Using Ensembl Resources genetic disorders
Phenotype Match
Exomiser NA Starts with phenotypes presented in HPO terms and a VCF file from [81]
WES/​WGS data, uses a Random Forest model to find likely causative
variants
PHIVE PHenotypic Interpretation of Conducts cross-​species phenotype matching and calculates phenotypic [69]
Variants in Exomes similarity between human diseases and animal models. Part of Exomiser
Phenolyzer Phenotype Based Gene Analyzer Prioritizes disease genes based on disease/​phenotype information [82]
entered as free text
Clinical Sequencing and Detection of Actionable Variants
Phenomizer NA Provides a semantic similarity search algorithm to statistically match a [72]
patient’s phenotypes to diseases. Also used by other tools such as Phen-​
Gen and PhenIX
Phevor NA Uses as input a list of variants generated from a variant ranking tool, [73]
reprioritizes the variants based on existing knowledge in multiple
biomedical ontologies of the affected genes, patient phenotype, and
human diseases
PhenIX Phenotypic Interpretation of Ranks variants in known disease genes based on similarity of patient [28]
eXomes phenotypes to phenotypes associated with the disease genes
eXtasy NA Uses a Random Forest classifier to rank missense variants based [70]
on patient phenotypes, predicted variant deleteriousness and
haploinsufficiency
Phen-​Gen NA Combines patient phenotypic and variant prediction data within a [74]
Bayesian framework to locate causative variants
DeepPVP Deep PhenomeNET Variant Identifies likely causative variants based on phenotype match using a [76]
Predictor deep neural network model

253
254 Next-Generation Sequencing Data Analysis

mutation spot affecting key functional domain of the coded protein, to those
presumed to be de novo; and (4) Supporting (PP) –​covers from those that
show co-​segregation with disease in multiple members of the affected family
in a gene known to cause the disease, to those that are called pathogenic by a
reputable source but with no available evidence.
For benign variants, the evidence of benign impact is divided into three
levels: (1) Stand-​Alone (coded as BA1) –​this level contains variants with
allele frequency above 5% in ESP, 1KGP, or gnomAD; (2) Strong (BS) –​covers
from those with allele frequency higher than expected for the disease to those
lacking segregation in the affected family; and (3) Supporting (BP) –​includes
missense variants in a gene where truncation variants are the primary mech-
anism of disease to synonymous variants with no evolutionary conservation
and no predicted effect on splicing.
For detailed definition and interpretation of the various levels of evidence,
and the rules for classifying variants into one of the five tiers based on different
combinations of these various evidence (summarized in Figure 11.2), readers
can refer to the original ACMG/​AMP publication ([89], particularly Tables 3,
4, and 5) and subsequent revisions. To follow the ACMG/​AMP guidelines,
all available evidence on the pathogenicity or benignity of a variant needs
to be considered. Such evidence includes those gathered from the current
case, data in public databases (such as those listed on Table 11.1) and sci-
entific literature, and/​or the clinal sequencing lab’s internal data. Through
reviewing the aggregated evidence and then applying the ACMG/​AMP
guidelines, the classification of variants can then be attained. Open-​source
or commercial tools, such as InterVar (Clinical Interpretation of Genetic
Variants, open source) or its web version wInterVar [90], QIAGEN Clinical
Insight (QCI) Interpret (commercial), may help classify variants by pro-
viding automated application of many of the criteria from the ACMG/​AMP
guidelines. CardioClassifier [91] and CardioVAI [92] are other examples of
decision support tools specifically developed for particular diseases, as both
aim to classify genes related to cardiac diseases. Prior to reporting, output
from decision support tools needs to be examined, and modified if necessary,
by testing lab personnel.

11.5.2 Classification of Somatic Variants


As the above ACMG/​AMP five-​tier system has been developed for germline
mutations in Mendelian disorders to classify somatic variants in tumor
samples, a different system has been proposed and recommended by AMP,
American Society of Clinical Oncology (ASCO), and College of American
Pathologists (CAP) [93]. This is a four-​tier system with emphasis on variants’
impact on clinical care including diagnosis, prognosis, therapy selection,
and inclusion in clinical trials. The four tiers are: Tier I –​variants of strong
clinical significance, Tier II –​variants of potential clinical significance, Tier
III –​those of unknown clinical significance, and Tier IV –​benign or likely
Clinical Sequencing and Detection of Actionable Variants 255

FIGURE 11.2
ACMG/​ AMP rules on how to classify variants based on combination of pathogenicity/​
benignity evidence at different levels. (From SE Brnich, EA Rivera-​Munoz, JS Berg, Quantifying
the potential of functional evidence to reclassify variants of uncertain significance in the
categorical and Bayesian interpretation frameworks, Human Mutation 2018, 39(11):1531–​1541.
With permission.)

benign variants. To categorize into one of these four tiers, four levels of evi-
dence are evaluated: (1) Level A –​variants that can serve as biomarkers to
predict response or resistance to approved therapies, or are included in pro-
fessional guidelines for a specific cancer type; (2) Level B –​those that can
256 Next-Generation Sequencing Data Analysis

predict response or resistance to therapies based on well-​powered studies


with expert consensus; (3) Level C –​those that can predict response or resist-
ance to approved therapies for a different tumor type, or qualify patients to
participate in clinical trials; (4) Level D –​those that can predict response or
resistance in multiple small trials with some consensus. To be classified into
Tier I, a variant needs to have Levels A and B evidence. For Tier II, a variant
needs to have Level C or D evidence. A Tier III variant is one that occurs in
a gene known to be cancer related, but how it is specifically associated with
cancer of any type has not been established. Variants that are observed in the
general population (or a specific subpopulation) at a frequency that is high
enough to preclude their connection with any cancer belong to Tier IV.
To help facilitate classification of oncogenic somatic variants, open-​source
or commercial tools can be used, such as VIC (Variant Interpretation for
Cancer, open source) [94], CancerVar (Cancer Variants interpretation, open
source) [95], Roche’s NAVIFY Mutation Profiler (commercial), and Qiagen’s
QCI Interpret (commercial). As an example, VIC, a Java-​based tool, applies
the AMP/​ASCO/​CAP classification rules to place somatic variants into one
of the tiers, based on assessment of their clinical impacts using information
from a number of databases including CGI (Cancer Genome Interpreter)
[96], CIViC (Clinical Interpretations of Variants in Cancer) [97], and PMKB
(Precision Medicine Knowledge Base) [98]. Following this automated process,
expert knowledge and patient-​specific case information can be incorporated
to adjust the final classification for reporting and utilization by clinical
oncologists and molecular tumor board for decision making.

11.6 Clinical Review and Reporting


11.6.1 Use of Artificial Intelligence in Variant Reporting
The use of AI in variant reporting can be exemplified by an effort from
Massachusetts General Hospital in which an AI system was built and validated
using variants that had been prioritized and reported by expert pathologists
from established patient cases [99]. To build machine learning models, this
system uses a large number of features, including the many mentioned above
such as variant frequency and prediction scores from CADD, SIFT, PolyPhen,
LRT, MutationTaster, MutationAssessor, FATHMM, PROVEAN, VEST,
phyloP, phastCons, GERP, SiPhy, etc. Machine learning algorithms used to
build these models included logistic regression and random forests among
others. This decision support tool generates a contiguous prediction score
between 0 (not reportable) to 1 (reportable) to help decide whether a variant
should be considered for reporting with human interpretable rationale. Other
commercially available AI-​based decision support tools, such as MOON [27],
Clinical Sequencing and Detection of Actionable Variants 257

are also available to assist with the variant reporting process. While they
cannot entirely replace clinical geneticists and pathologists, these AI-​based
decision support tools can help filter out most non-​reportable variants to
allow experts to focus on a more manageable number of potentially report-
able variants, making this key analytic process more efficient.

11.6.2 Expert Review
While the variant reporting process can be automated with promising results
using the tools introduced above, review of reportable pathogenic or likely
pathogenic germline variants, or Tier I or II somatic variants, by molecular
pathologists and oncologists is still needed to examine the entire evidence
matrix for their pathogenic/​oncogenic role prior to reporting. Current med-
ical literature and database entries should be checked for their links with
known diseases or implicated biological pathways. This step should also be
contextualized in consideration of the patient’s phenotype and family his-
tory. Such expert manual review is needed to avoid non-​pathogenic/​non-​
oncogenic variants from being reported as false positives.

11.6.3 Generation of Testing Report


After expert review, a final clinical report needs to be prepared by the testing
lab to convey the results to clinicians for decision making. While there is
no standard format for such a report, as a general rule the report should be
designed toward helping with bedside decision making, and therefore needs to
be concise, informative, actionable, and easy to understand to non-​geneticists
(Figure 11.3 shows an example). At the beginning of the report, an executive
summary of major findings should be provided succinctly, followed by more
detailed interpretations of the variants. Variants in the major findings may
be limited only to those that are classified as pathogenic or likely pathogenic,
but some labs may opt to also include variants of unknown significance.
Typically variants that are benign or likely benign are not reported. It is pos-
sible to report a negative finding, i.e., no genetic cause of the patient’s pheno-
type is identified. On the report the variants and the genes harboring them
should be described using the nomenclature from Human Genome Variation
Society (HGVS) and Human Genome Organization Gene Nomenclature
Committee (HGNC) [100, 101]. Each variant should be presented with a
reference sequence, a letter prefix (such as “c” for a DNA coding reference
sequence, “p” for a protein reference, or “r” for an RNA transcript sequence),
description of the sequence change at a DNA/​RNA/​protein sequence loca-
tion, zygosity, medical condition, inheritance pattern, and classification (e.g.,
ABCA4, NM_​000350.2, c.5882G>A, p.Gly1961Glu, heterozygous, age-​related
macular degeneration, autosomal dominant, likely pathogenic). A detailed
description for each of the identified variants may include evidence level(s)
newgenrtpdf
258
Next-Generation Sequencing Data Analysis
FIGURE 11.3
Clinical sequencing testing report example. (Adapted from: GD Farmer, H Gray, G Chandratillake, FL Raymond, ALJ Freeman, Recommendations for
designing genetic test reports to be understood by patients and non-​specialists, European Journal of Human Genetics 2020, 28(7):885. Used under the terms
of the Creative Commons Attribution 4.0 International License, https://​crea​tive​comm​ons.org/​licen​ses/​by/​4.0, ©2020 Farmer et al.)
Clinical Sequencing and Detection of Actionable Variants 259

for the classification of the variant, literature summary of disease/​phenotype


connection, and other key information supporting the variant’s pathogen-
icity. This can be followed by recommendations for further actions, such as
strategies for intervention and/​or patient management including prognosis
and diagnosis. Such recommendations need to be based on evidence, and
considered in the context of the patient’s specific condition. If a clinical trial
is available, e.g., for a cancer patient carrying a Tier I somatic variant, this
information should also be included in the report.
Other sections of the clinical report should also contain technical informa-
tion about the test, including what gene or genome content was assayed, how
the test was performed, as well as other technological factors such as the
genome build used for analysis that may have an impact on test result gen-
eration and interpretation. Such factors include limit of detection, minimum
sequencing coverage, actual test performance metrics, etc. Limitations of the
test, such as low-​coverage and non-​targeted genomic regions, should also
be made clear. To provide further reference, literature citations or links to
database sites should be provided toward the end. Additional clinically rele-
vant and actionable information may also be provided from resources such
as GeneReviews, which provides a quick reference to clinicians on hereditary
disorders. For patients and their family members, a section that cites easy-​
to-​understand genetic health information from additional resources, such as
MedlinePlus Genetics, may also be helpful.

11.6.4 Variant Validation
While traditionally germline SNVs or small indels on a clinical report need to
be validated using an orthogonal technology, with Sanger sequencing being
the golden standard, a number of studies have shown that this is not always
necessary and NGS is often more accurate than Sanger sequencing [102, 103].
CNVs are typically validated using orthogonal techniques such as multiplex
ligation-​
dependent probe amplification (MLPA) or comparative genomic
hybridization arrays (aCGH), but validation using an NGS CNV pipeline
has been reported [104]. Somatic variants detected in cancerous samples, on
the other hand, should be validated using an orthogonal method, including
Sanger sequencing, digital PCR, or pyrosequencing, with the latter two
especially suited for those detected at low frequencies. It should be noted
again that for both germline and somatic variants, the preliminary variant
screening step mentioned earlier to filter out low-​ quality variants is an
important step to minimize the rate of false-​positive variants. In addition, to
ensure call accuracy of the variants on the final report, their raw reads should
be manually examined by visually checking pileup of reads that align to their
genomic locations using a visualization tool such as Integrative Genomics
Viewer (IGV). This simple step can help further reduce false positives caused
by factors such as insufficient sequencing coverage. The use of these QC
measures, as well as sound medical judgment and good clinical practice, are
260 Next-Generation Sequencing Data Analysis

fundamental to generating a report that captures representative variants in


the patient. How the reported variants are validated or confirmed should
also be included in the report.

11.6.5 Incorporation into a Patient’s Electronic Health Record


A patient clinical sequencing report is increasingly incorporated into the
patient’s EHR. While the report can be entered into EHR as an unstructured
PDF, for better integration with the rest of EHR, new data standards and
interfaces need to be developed. Currently, Health Level Seven International
(HL7) and Global Alliance for Genomics and Health (GA4GH) are developing
new data models and data exchange standards to facilitate collection,
coding, and retrieval of clinical genomics data. HL7, for example, establishes
standards for EHR data exchange, integration, and retrieval that are widely
accepted and used by clinical computer systems in hospitals and other health
care-​related organizations. Fast Healthcare Interoperability Resources (FHIR)
Genomics develops and provides implementation of the HL7 data standards
for clinical genomics data. Incorporation of patient sequencing variant
report into EHR using such standards provides future-​proof clinical decision
support and analytics.

11.6.6 Reporting of Secondary Findings


When performing WGS/​WES, besides reporting variants related to the indi-
cation that leads to the ordering of clinical sequencing, ACMG recommends
to also report Pathogenic or Likely Pathogenic variants in a short list of med-
ically significant but actionable genes. The genes on this list are known to
lead to disease phenotypes, such as cancer and cardiovascular phenotypes,
that have severe medical implications and high lifetime penetrance. The goal
of reporting such variants, termed secondary finding (previously called inci-
dental finding, or unsolicited finding), is to help improve health by reducing
morbidity and mortality through taking appropriate actions. The current list
(ACMG SF v3.0) [105] contains 73 genes, but the number is bound to change
with the addition of new genes or removal of existing genes. Based on the
current ACMG recommendations [106], patients and family members should
be consented on receiving secondary findings with the option to opt out.

11.6.7 Patient Counseling and Periodic Report Updates


To provide interpretation to the patient (and their family members) on primary
as well as secondary findings, and help them understand the implications of
such findings, genetic counseling is needed. On primary findings, the role
of genetic counselors is to help patients and families better understand the
testing results and treatment options, as well as limitations of the test. On
secondary findings, the counseling includes providing interpretation on the
Clinical Sequencing and Detection of Actionable Variants 261

probability of developing the indicated disorder(s), strategies to prevent or


manage the indicated risk(s), as well as support on emotional concerns and
referral to other health providers and support groups. In addition to post-​test
counseling, genetic counselors also provide pre-​testing counseling for indi-
viduals who have symptoms of a particular genetic disorder, are likely to
be affected by a genetic disorder because of family history, or are concerned
about transmitting a condition to the next generation for the purpose of
family planning.
Variant classification is a dynamic process. With accumulation of new evi-
dence for gene and variant pathogenicity over time, it frequently becomes
necessary to reclassify variants on the original report, for example, from
Unknown Significance to Likely Pathogenic or Likely Benign, or less fre-
quently from Likely Pathogenic to Benign, etc. In addition, new patient
phenotypic data (e.g., new symptoms) and/​or new family history informa-
tion, or reports of other new cases that share similar symptoms or variants,
may also prompt reanalysis of the original sequencing data. Furthermore,
advancement in bioinformatics tool development provides another impetus
for reanalysis. Besides improving variant classification, newer tools may also
lead to identification of variants that were not found initially. Because of such
advances in knowledge generation, patient phenotyping, and tool devel-
opment, it is beneficial to periodically update the original report in order
to provide new guidance that would impact patient clinical management.
Performance of reclassification of variants on the original report and/​or
reanalysis of the original data to provide periodic updates, however, requires
time and resources from testing labs, clinicians, and genetic counselors. Such
longevity report updates may also have long-​term implications, including
elicitation of psychological distress to patients and their families, as a
result some may prefer not to be recontacted. While the ethical and legal
implications of reinterpretation/​reanalysis/​recontact are still being debated
[107, 108], current guidelines from professional organizations such as ACMG
focus on maximizing clinical impact of providing such updates while minim-
izing the burden on the test lab and health care system [109].

11.7 Bioinformatics Pipeline Validation


Besides wet lab workflow, a dry lab bioinformatics pipeline must also be
rigorously tested and validated before deployment. This is to meet accredit-
ation requirements by professional organizations such as CAP, and more
importantly, to ensure that all variables that might affect specificity, sensi-
tivity, and precision/​reproducibility of a test are well defined and valid. On
bioinformatics pipeline validation, AMP and CAP have published detailed
standards and guidelines [110], which are summarized below. Validation is
262 Next-Generation Sequencing Data Analysis

needed not only for labs that develop their own custom steps, but also for
those that opt to use a commercially available platform. In the latter case,
although the service provider may offer a pre-​validated platform, the testing
lab still needs to go through familiarization and optimization, and validate
that the platform meets the designed analytical goals of the test. To help val-
idate a new or existing pipeline and evaluate its performance, the reference
standard samples used for validating the lab workflow (see Section 11.1.2),
including the GIAB reference DNA and bio-​engineered DNA that contains
synthetic variants at predefined frequencies, are equally valuable here since
they provide the ground truth. In addition, bioinformatically generated ref-
erence dataset using programs such as BAMSurgeon [111] may also be used.
For the validation, all analytic steps in the pipeline need to be clearly
defined with required hardware and software specified for each step. Such
specifics include hardware configuration, operating system, name and
version number of software and their dependencies, data storage and trans-
mission system, and network connection protocol. In addition, other analytic
details such as parameters used in each software, reference genome used for
alignment, and databases accessed for annotation and filtering, should also
be specified. Any sequencing reads altering operations, such as trimming,
should be fully evaluated to determine whether they are appropriate, or need
to be revised or dropped. Quality metrics for each of the analytic steps, such
as mean reads on-​target coverage and percentage of target genomic regions
with coverage over a threshold (for reads alignment), and depth of coverage
for each called variant (variant calling), should be compared to pre-​defined
performance criteria. Applied variant filters should be evaluated carefully to
make sure that true positives are not filtered out.
If the pipeline uses internally developed software tools and scripts, the com-
puter code should be deposited in a source code repository, such as GitHub,
BitBucket, or SourceForge. If using externally developed software, the source
code should also be documented if accessible. Also as part of the validation
process, the strategies established to back up data and maintain the integ-
rity of raw and analyzed files during transfer should be evaluated. On the
issue of legal compliance, the pipeline should follow all applicable laws at
the national and local levels. In the United States, the applicable laws include
the Health Insurance Portability and Accountability Act (HIPAA) and other
national/​state/​local laws that pertain to clinical genetic testing. According
to these laws, patient genetic information needs to be protected like other
patient information including patient identity and other health records, and
should be secured throughout the analytic process. When using a commer-
cial system, it is the responsibility of the testing lab to verify such compliance
issues. To avoid accidental mixing of patient data, based on the AMP/​CAP
guidelines the identity of a patient sample must be preserved throughout the
analytic process using at least four unique identifiers to encompass sample,
patient, run, and test location [110]. As all analytic pipelines have limitations,
such limitations for a validated pipeline should be clearly documented
Clinical Sequencing and Detection of Actionable Variants 263

and reported. After a pipeline is validated, if there is any update or change


applied to any part of the pipeline, supplemental or new validation is needed
depending on the scope of the update/​change.

References
1. Patel LR, Nykter M, Chen K, Zhang W. Cancer genome sequen-
cing: understanding malignancy as a disease of the genome, its conformation,
and its evolution. Cancer letters 2013, 340(2):152–​160.
2. Steuer CE, Ramalingam SS. Tumor mutation burden: leading immunotherapy
to the era of precision medicine? J Clin Oncol 2018, 36(7):631–​632.
3. Farnaes L, Hildreth A, Sweeney NM, Clark MM, Chowdhury S, Nahas
S, Cakici JA, Benson W, Kaplan RH, Kronick R et al. Rapid whole-​genome
sequencing decreases infant morbidity and cost of hospitalization. NPJ Genom
Med 2018, 3:10.
4. Jones S, Anagnostou V, Lytle K, Parpart-​ Li S, Nesselbush M, Riley DR,
Shukla M, Chesnick B, Kadan M, Papp E et al. Personalized genomic ana-
lyses for cancer mutation discovery and interpretation. Sci Transl Med 2015,
7(283):283ra253.
5. Srinivasan M, Sedmak D, Jewell S. Effect of fixatives and tissue pro-
cessing on the content and integrity of nucleic acids. Am J Pathol 2002,
161(6):1961–​1971.
6. Hedegaard J, Thorsen K, Lund MK, Hein AM, Hamilton-​Dutoit SJ, Vang S,
Nordentoft I, Birkenkamp-​Demtroder K, Kruhoffer M, Hager H et al. Next-​
generation sequencing of RNA and DNA isolated from paired fresh-​frozen
and formalin-​fixed paraffin-​embedded samples of human cancer and normal
tissue. PLoS One 2014, 9(5):e98187.
7. Do H, Dobrovic A. Sequence artifacts in DNA from formalin-​ fixed
tissues: causes and strategies for minimization. Clin Chem 2015, 61(1):64–​71.
8. McDonough SJ, Bhagwate A, Sun ZF, Wang C, Zschunke M, Gorman JA, Kopp
KJ, Cunningham JM. Use of FFPE-​derived DNA in next generation sequen-
cing: DNA extraction methods. Plos One 2019, 14(4).
9. Oreskovic A, Brault ND, Panpradist N, Lai JJ, Lutz BR. Analytical comparison
of methods for extraction of short cell-​free DNA from urine. J Mol Diagn 2019,
21(6):1067–​1078.
10. Diefenbach RJ, Lee JH, Kefford RF, Rizos H. Evaluation of commercial kits for
purification of circulating free DNA. Cancer Genet 2018, 228–​229:21–​27.
11. Alborelli I, Generali D, Jermann P, Cappelletti MR, Ferrero G, Scaggiante B,
Bortul M, Zanconati F, Nicolet S, Haegele J et al. Cell-​free DNA analysis in
healthy individuals by next-​generation sequencing: a proof of concept and
technical validation study. Cell Death Dis 2019, 10(7):534.
12. Jiang P, Chan CW, Chan KC, Cheng SH, Wong J, Wong VW, Wong GL,
Chan SL, Mok TS, Chan HL et al. Lengthening and shortening of plasma
DNA in hepatocellular carcinoma patients. Proc Natl Acad Sci U S A 2015,
112(11):E1317–​1325.
264 Next-Generation Sequencing Data Analysis

13. Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, Foye A, Lloyd P,
Nykter M, Beer TM et al. Concordance of circulating tumor DNA and matched
metastatic tissue biopsy in prostate cancer. J Natl Cancer Inst 2017, 109(12).
14. Chen M, Zhao H. Next-​ generation sequencing in liquid biopsy: cancer
screening and early detection. Hum Genomics 2019, 13(1):34.
15. Jones AG, Small CM, Paczolt KA, Ratterman NL. A practical guide to methods
of parentage analysis. Mol Ecol Resour 2010, 10(1):6–​30.
16. Zhang L, Dong X, Lee M, Maslov AY, Wang T, Vijg J. Single-​cell whole-​
genome sequencing reveals the functional landscape of somatic mutations
in B lymphocytes across the human lifespan. Proc Natl Acad Sci U S A 2019,
116(18):9014–​9019.
17. Petrackova A, Vasinek M, Sedlarikova L, Dyskova T, Schneiderova P, Novosad
T, Papajik T, Kriegova E. Standardization of sequencing coverage depth in
NGS: recommendation for detection of clonal and subclonal mutations in
cancer diagnostics. Front Oncol 2019, 9:851.
18. Meggendorfer M, Jobanputra V, Wrzeszczynski KO, Roepman P, de Bruijn E,
Cuppen E, Buttner R, Caldas C, Grimmond S, Mullighan CG et al. Analytical
demands to use whole-​ genome sequencing in precision oncology. Semin
Cancer Biol 2021, 84:16–​22.
19. Salk JJ, Schmitt MW, Loeb LA. Enhancing the accuracy of next-​generation
sequencing for detecting rare and subclonal mutations. Nat Rev Genet 2018,
19(5):269–​285.
20. Schmitt MW, Kennedy SR, Salk JJ, Fox EJ, Hiatt JB, Loeb LA. Detection of
ultra-​rare mutations by next-​generation sequencing. Proc Natl Acad Sci U S A
2012, 109(36):14508–​14513.
21. Zook JM, McDaniel J, Olson ND, Wagner J, Parikh H, Heaton H, Irvine SA, Trigg
L, Truty R, McLean CY et al. An open resource for accurately benchmarking
small variant and reference calls. Nat Biotechnol 2019, 37(5):561–​566.
22. Miller NA, Farrow EG, Gibson M, Willig LK, Twist G, Yoo B, Marrs T, Corder
S, Krivohlavek L, Walter A et al. A 26-​hour system of highly sensitive whole
genome sequencing for emergency management of genetic diseases. Genome
Med 2015, 7(1):100.
23. Mestek-​Boukhibar L, Clement E, Jones WD, Drury S, Ocaka L, Gagunashvili
A, Le Quesne Stabej P, Bacchelli C, Jani N, Rahman S et al. Rapid Paediatric
Sequencing (RaPS): comprehensive real-​life workflow for rapid diagnosis of
critically ill children. J Med Genet 2018, 55(11):721–​728.
24. Kendig KI, Baheti S, Bockol MA, Drucker TM, Hart SN, Heldenbrand JR,
Hernaez M, Hudson ME, Kalmbach MT, Klee EW et al. Sentieon DNASeq
variant calling workflow demonstrates strong computational performance
and accuracy. Front Genet 2019, 10:736.
25. Loka TP, Tausch SH, Renard BY. Reliable variant calling during runtime of
Illumina sequencing. Sci Rep 2019, 9(1):16502.
26. Stranneheim H, Engvall M, Naess K, Lesko N, Larsson P, Dahlberg M, Andeer
R, Wredenberg A, Freyer C, Barbaro M et al. Rapid pulsed whole genome
sequencing for comprehensive acute diagnostics of inborn errors of metab-
olism. BMC Genomics 2014, 15:1090.
27. Clark MM, Hildreth A, Batalov S, Ding Y, Chowdhury S, Watkins K, Ellsworth
K, Camp B, Kint CI, Yacoubian C et al. Diagnosis of genetic diseases in seriously
Clinical Sequencing and Detection of Actionable Variants 265

ill children by rapid whole-​genome sequencing and automated phenotyping


and interpretation. Sci Transl Med 2019, 11(489):eaat6177.
28. Zemojtel T, Kohler S, Mackenroth L, Jager M, Hecht J, Krawitz P, Graul-​
Neumann L, Doelken S, Ehmke N, Spielmann M et al. Effective diagnosis of
genetic disease by computational phenotype analysis of the disease-​associated
genome. Sci Transl Med 2014, 6(252):252ra123.
29. Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J, Wang Q,
Collins RL, Laricchia KM, Ganna A, Birnbaum DP et al. The mutational con-
straint spectrum quantified from variation in 141,456 humans. Nature 2020,
581(7809):434–​443.
30. Siva N. 1000 Genomes project. Nat Biotechnol 2008, 26(3):256.
31. Taliun D, Harris DN, Kessler MD, Carlson J, Szpiech ZA, Torres R, Taliun
SAG, Corvelo A, Gogarten SM, Kang HM et al. Sequencing of 53,831 diverse
genomes from the NHLBI TOPMed Program. Nature 2021, 590(7845):290–​299.
32. Consortium UK, Walter K, Min JL, Huang J, Crooks L, Memari Y, McCarthy S,
Perry JR, Xu C, Futema M et al. The UK10K project identifies rare variants in
health and disease. Nature 2015, 526(7571):82–​90.
33. Exome Variant Server, NHLBI GO Exome Sequencing Project (ESP) (http://​
evs.gs.was​hing​ton.edu/​EVS/​)
34. MacDonald JR, Ziman R, Yuen RK, Feuk L, Scherer SW. The Database of
Genomic Variants: a curated collection of structural variation in the human
genome. Nucleic Acids Res 2014, 42(Database issue):D986–​992.
35. Firth HV, Richards SM, Bevan AP, Clayton S, Corpas M, Rajan D, Van Vooren
S, Moreau Y, Pettett RM, Carter NP. DECIPHER: Database of Chromosomal
Imbalance and Phenotype in Humans Using Ensembl Resources. Am J Hum
Genet 2009, 84(4):524–​533.
36. Lappalainen I, Lopez J, Skipper L, Hefferon T, Spalding JD, Garner J, Chen C,
Maguire M, Corbett M, Zhou G et al. DbVar and DGVa: public archives for gen-
omic structural variation. Nucleic Acids Res 2013, 41(Database issue):D936–​941.
37. Obenchain V, Lawrence M, Carey V, Gogarten S, Shannon P, Morgan M.
VariantAnnotation: a Bioconductor package for exploration and annotation of
genetic variants. Bioinformatics 2014, 30(14):2076–​2078.
38. Jaganathan K, Kyriazopoulou Panagiotopoulou S, McRae JF, Darbandi SF,
Knowles D, Li YI, Kosmicki JA, Arbelaez J, Cui W, Schwartz GB et al. Predicting
splicing from primary sequence with deep learning. Cell 2019, 176(3):535–​
548 e524.
39. Yeo G, Burge CB. Maximum entropy modeling of short sequence motifs with
applications to RNA splicing signals. J Comput Biol 2004, 11(2–​3):377–​394.
40. Reese MG, Eeckman FH, Kulp D, Haussler D. Improved splice site detection
in Genie. J Comput Biol 1997, 4(3):311–​323.
41. Jian X, Boerwinkle E, Liu X. In silico prediction of splice-​altering single nucleo-
tide variants in the human genome. Nucleic Acids Res 2014, 42(22):13534–​13544.
42. Wai HA, Lord J, Lyon M, Gunning A, Kelly H, Cibin P, Seaby EG, Spiers-​
Fitzgerald K, Lye J, Ellard S et al. Blood RNA analysis can increase clinical
diagnostic rate and resolve variants of uncertain significance. Genet Med 2020,
22(6):1005–​1014.
43. Riepe TV, Khan M, Roosing S, Cremers FPM, t Hoen PAC. Benchmarking deep
learning splice prediction tools using functional splice assays. Hum Mutat
2021, 42(7):799–​810.
266 Next-Generation Sequencing Data Analysis

44. Rentzsch P, Schubach M, Shendure J, Kircher M. CADD-​Splice-​improving


genome-​wide variant effect prediction using deep learning-​derived splice
scores. Genome Med 2021, 13(1):31.
45. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P,
Kondrashov AS, Sunyaev SR. A method and server for predicting damaging
missense mutations. Nat Methods 2010, 7(4):248–​249.
46. Choi Y, Chan AP. PROVEAN web server: a tool to predict the functional effect
of amino acid substitutions and indels. Bioinformatics 2015, 31(16):2745–​2747.
47. Vaser R, Adusumalli S, Leng SN, Sikic M, Ng PC. SIFT missense predictions
for genomes. Nat Protoc 2016, 11(1):1–​9.
48. Steinhaus R, Proft S, Schuelke M, Cooper DN, Schwarz JM, Seelow D.
MutationTaster2021. Nucleic Acids Res 2021, 49(W1):W446–​W451.
49. Reva B, Antipin Y, Sander C. Predicting the functional impact of protein
mutations: application to cancer genomics. Nucleic Acids Res 2011, 39(17):e118.
50. Carter H, Douville C, Stenson PD, Cooper DN, Karchin R. Identifying
Mendelian disease genes with the variant effect scoring tool. BMC Genomics
2013, 14 Suppl 3:S3.
51. Shihab HA, Gough J, Cooper DN, Stenson PD, Barker GL, Edwards KJ, Day IN,
Gaunt TR. Predicting the functional, molecular, and phenotypic consequences
of amino acid substitutions using hidden Markov models. Hum Mutat 2013,
34(1):57–​65.
52. Pejaver V, Urresti J, Lugo-​Martinez J, Pagel KA, Lin GN, Nam HJ, Mort M,
Cooper DN, Sebat J, Iakoucheva LM et al. Inferring the molecular and pheno-
typic impact of amino acid variants with MutPred2. Nat Commun 2020,
11(1):5918.
53. Chun S, Fay JC. Identification of deleterious mutations within three human
genomes. Genome Res 2009, 19(9):1553–​1561.
54. Siepel A, Bejerano G, Pedersen JS, Hinrichs AS, Hou M, Rosenbloom K,
Clawson H, Spieth J, Hillier LW, Richards S et al. Evolutionarily conserved
elements in vertebrate, insect, worm, and yeast genomes. Genome Res 2005,
15(8):1034–​1050.
55. Cooper GM, Stone EA, Asimenos G, Program NCS, Green ED, Batzoglou S,
Sidow A. Distribution and intensity of constraint in mammalian genomic
sequence. Genome Res 2005, 15(7):901–​913.
56. Garber M, Guttman M, Clamp M, Zody MC, Friedman N, Xie X. Identifying
novel constrained elements by exploiting biased substitution patterns.
Bioinformatics 2009, 25(12):i54–​62.
57. Rentzsch P, Witten D, Cooper GM, Shendure J, Kircher M. CADD: predicting
the deleteriousness of variants throughout the human genome. Nucleic Acids
Res 2019, 47(D1):D886–​D894.
58. Jagadeesh KA, Wenger AM, Berger MJ, Guturu H, Stenson PD, Cooper
DN, Bernstein JA, Bejerano G. M-​CAP eliminates a majority of variants of
uncertain significance in clinical exomes at high sensitivity. Nat Genet 2016,
48(12):1581–​1586.
59. Li C, Zhi D, Wang K, Liu X. MetaRNN: Differentiating Rare Pathogenic and
Rare Benign Missense SNVs and InDels using deep learning. bioRxiv 2021.
60. Ioannidis NM, Rothstein JH, Pejaver V, Middha S, McDonnell SK, Baheti S,
Musolf A, Li Q, Holzinger E, Karyadi D et al. REVEL: an ensemble method for
Clinical Sequencing and Detection of Actionable Variants 267

predicting the pathogenicity of rare missense variants. Am J Hum Genet 2016,


99(4):877–​885.
61. Ionita-​Laza I, McCallum K, Xu B, Buxbaum JD. A spectral approach inte-
grating functional genomic annotations for coding and noncoding variants.
Nat Genet 2016, 48(2):214–​220.
62. Hu H, Huff CD, Moore B, Flygare S, Reese MG, Yandell M. VAAST
2.0: improved variant classification and disease-​gene identification using a
conservation-​controlled amino acid substitution matrix. Genet Epidemiol 2013,
37(6):622–​634.
63. Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, Gu B, Hart
J, Hoffman D, Jang W et al. ClinVar: improving access to variant interpret-
ations and supporting evidence. Nucleic Acids Res 2018, 46(D1):D1062–​D1067.
64. Stenson PD, Ball EV, Mort M, Phillips AD, Shaw K, Cooper DN. The Human
Gene Mutation Database (HGMD) and its exploitation in the fields of
personalized genomics and molecular evolution. Curr Protoc Bioinformatics
2012, Chapter 1:1.13.1-​1.13.20.
65. Online Mendelian Inheritance in Man, OMIM (https://​omim.org/​)
66. Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, Boutselakis
H, Cole CG, Creatore C, Dawson E et al. COSMIC: the Catalogue Of Somatic
Mutations In Cancer. Nucleic Acids Res 2019, 47(D1):D941–​D947.
67. Grossman RL, Heath AP, Ferretti V, Varmus HE, Lowy DR, Kibbe WA, Staudt
LM. Toward a Shared Vision for Cancer Genomic Data. N Engl J Med 2016,
375(12):1109–​1112.
68. Consortium APG. AACR Project GENIE: powering precision medicine
through an International Consortium. Cancer Discov 2017, 7(8):818–​831.
69. Robinson PN, Kohler S, Oellrich A, Sanger Mouse Genetics P, Wang K, Mungall
CJ, Lewis SE, Washington N, Bauer S, Seelow D et al. Improved exome pri-
oritization of disease genes through cross-​species phenotype comparison.
Genome Res 2014, 24(2):340–​348.
70. Sifrim A, Popovic D, Tranchevent LC, Ardeshirdavani A, Sakai R, Konings P,
Vermeesch JR, Aerts J, De Moor B, Moreau Y. eXtasy: variant prioritization by
genomic data fusion. Nat Methods 2013, 10(11):1083–​1084.
71. Smedley D, Schubach M, Jacobsen JOB, Kohler S, Zemojtel T, Spielmann M,
Jager M, Hochheiser H, Washington NL, McMurry JA et al. A whole-​genome
analysis framework for effective identification of pathogenic regulatory
variants in Mendelian disease. Am J Hum Genet 2016, 99(3):595–​606.
72. Kohler S, Schulz MH, Krawitz P, Bauer S, Dolken S, Ott CE, Mundlos C, Horn
D, Mundlos S, Robinson PN. Clinical diagnostics in human genetics with
semantic similarity searches in ontologies. Am J Hum Genet 2009, 85(4):457–​464.
73. Singleton MV, Guthery SL, Voelkerding KV, Chen K, Kennedy B, Margraf
RL, Durtschi J, Eilbeck K, Reese MG, Jorde LB et al. Phevor combines mul-
tiple biomedical ontologies for accurate identification of disease-​ causing
alleles in single individuals and small nuclear families. Am J Hum Genet 2014,
94(4):599–​610.
74. Javed A, Agrawal S, Ng PC. Phen-​Gen: combining phenotype and genotype to
analyze rare disorders. Nat Methods 2014, 11(9):935–​937.
75. Stelzer G, Plaschkes I, Oz-​Levi D, Alkelai A, Olender T, Zimmerman S, Twik
M, Belinky F, Fishilevich S, Nudel R et al. VarElect: the phenotype-​based vari-
ation prioritizer of the GeneCards Suite. BMC Genomics 2016, 17 Suppl 2:444.
268 Next-Generation Sequencing Data Analysis

76. Boudellioua I, Kulmanov M, Schofield PN, Gkoutos GV, Hoehndorf R.


DeepPVP: phenotype-​based prioritization of causative variants using deep
learning. BMC Bioinformatics 2019, 20(1):65.
77. Rodriguez-​Garcia MA, Gkoutos GV, Schofield PN, Hoehndorf R. Integrating
phenotype ontologies with PhenomeNET. J Biomed Semantics 2017, 8(1):58.
78. Davydov EV, Goode DL, Sirota M, Cooper GM, Sidow A, Batzoglou S.
Identifying a high fraction of the human genome to be under selective con-
straint using GERP+​+​. PLoS Comput Biol 2010, 6(12):e1001025.
79. Landrum MJ, Chitipiralla S, Brown GR, Chen C, Gu B, Hart J, Hoffman D,
Jang W, Kaur K, Liu C et al. ClinVar: improvements to accessing data. Nucleic
Acids Res 2020, 48(D1):D835–​D844.
80. Bragin E, Chatzimichali EA, Wright CF, Hurles ME, Firth HV, Bevan AP,
Swaminathan GJ. DECIPHER: database for the interpretation of phenotype-​
linked plausibly pathogenic sequence and copy-​number variation. Nucleic
Acids Res 2014, 42(Database issue):D993–​D1000.
81. Smedley D, Jacobsen JO, Jager M, Kohler S, Holtgrewe M, Schubach M,
Siragusa E, Zemojtel T, Buske OJ, Washington NL et al. Next-​generation
diagnostics and disease-​gene discovery with the Exomiser. Nat Protoc 2015,
10(12):2004–​2015.
82. Yang H, Robinson PN, Wang K. Phenolyzer: phenotype-​based prioritization
of candidate genes for human diseases. Nat Methods 2015, 12(9):841–​843.
83. Holtgrewe M, Stolpe O, Nieminen M, Mundlos S, Knaus A, Kornak U, Seelow
D, Segebrecht L, Spielmann M, Fischer-​Zirnsak B et al. VarFish: comprehen-
sive DNA variant analysis for diagnostics and research. Nucleic Acids Res 2020,
48(W1):W162–​W169.
84. Li MX, Gui HS, Kwan JS, Bao SY, Sham PC. A comprehensive framework
for prioritizing variants in exome sequencing studies of Mendelian diseases.
Nucleic Acids Res 2012, 40(7):e53.
85. Flygare S, Hernandez EJ, Phan L, Moore B, Li M, Fejes A, Hu H, Eilbeck K,
Huff C, Jorde L et al. The VAAST Variant Prioritizer (VVP): ultrafast, easy
to use whole genome variant prioritization tool. BMC Bioinformatics 2018,
19(1):57.
86. Geoffroy V, Pizot C, Redin C, Piton A, Vasli N, Stoetzel C, Blavier A, Laporte
J, Muller J. VaRank: a simple and powerful tool for ranking genetic variants.
PeerJ 2015, 3:e796.
87. Alexander J, Mantzaris D, Georgitsi M, Drineas P, Paschou P. Variant
Ranker: a web-​tool to rank genomic data according to functional significance.
BMC Bioinformatics 2017, 18(1):341.
88. Ip E, Chapman G, Winlaw D, Dunwoodie SL, Giannoulatou E. VPOT: A
Customizable Variant Prioritization Ordering Tool for Annotated Variants.
Genomics Proteomics Bioinformatics 2019, 17(5):540–​545.
89. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-​Foster J, Grody WW, Hegde
M, Lyon E, Spector E et al. Standards and guidelines for the interpretation
of sequence variants: a joint consensus recommendation of the American
College of Medical Genetics and Genomics and the Association for Molecular
Pathology. Genet Med 2015, 17(5):405–​424.
90. Li Q, Wang K. InterVar: Clinical Interpretation of Genetic Variants by the 2015
ACMG-​AMP Guidelines. Am J Hum Genet 2017, 100(2):267–​280.
Clinical Sequencing and Detection of Actionable Variants 269

91. Whiffin N, Walsh R, Govind R, Edwards M, Ahmad M, Zhang X, Tayal U,


Buchan R, Midwinter W, Wilk AE et al. CardioClassifier: disease-​and gene-​
specific computational decision support for clinical genome interpretation.
Genet Med 2018, 20(10):1246–​1254.
92. Nicora G, Limongelli I, Gambelli P, Memmi M, Malovini A, Mazzanti A,
Napolitano C, Priori S, Bellazzi R. CardioVAI: An automatic implementation
of ACMG-​AMP variant interpretation guidelines in the diagnosis of cardio-
vascular diseases. Hum Mutat 2018, 39(12):1835–​1846.
93. Li MM, Datto M, Duncavage EJ, Kulkarni S, Lindeman NI, Roy S,
Tsimberidou AM, Vnencak-​Jones CL, Wolff DJ, Younes A et al. Standards
and Guidelines for the Interpretation and Reporting of Sequence Variants in
Cancer: A Joint Consensus Recommendation of the Association for Molecular
Pathology, American Society of Clinical Oncology, and College of American
Pathologists. J Mol Diagn 2017, 19(1):4–​23.
94. He MM, Li Q, Yan M, Cao H, Hu Y, He KY, Cao K, Li MM, Wang K. Variant
Interpretation for Cancer (VIC): a computational tool for assessing clinical
impacts of somatic variants. Genome Med 2019, 11(1):53.
95. Li Q, Ren Z, Cao K, Li MM, Wang K, Zhou Y. CancerVar: an artificial
intelligence-​
empowered platform for clinical interpretation of somatic
mutations in cancer. Sci Adv 2022, 8(18):eabj1624.
96. Tamborero D, Rubio-​ Perez C, Deu-​ Pons J, Schroeder MP, Vivancos A,
Rovira A, Tusquets I, Albanell J, Rodon J, Tabernero J et al. Cancer Genome
Interpreter annotates the biological and clinical relevance of tumor alter-
ations. Genome Med 2018, 10(1):25.
97. Griffith M, Spies NC, Krysiak K, McMichael JF, Coffman AC, Danos AM,
Ainscough BJ, Ramirez CA, Rieke DT, Kujan L et al. CIViC is a commu-
nity knowledgebase for expert crowdsourcing the clinical interpretation of
variants in cancer. Nat Genet 2017, 49(2):170–​174.
98. Huang L, Fernandes H, Zia H, Tavassoli P, Rennert H, Pisapia D, Imielinski
M, Sboner A, Rubin MA, Kluk M et al. The cancer precision medicine know-
ledge base for structured clinical-​grade mutations and interpretations. J Am
Med Inform Assoc 2017, 24(3):513–​519.
99. Zomnir MG, Lipkin L, Pacula M, Meneses ED, MacLeay A, Duraisamy S,
Nadhamuni N, Al Turki SH, Zheng Z, Rivera M et al. Artificial intelligence
approach for variant reporting. JCO Clin Cancer Inform 2018, 2.
100. den Dunnen JT, Dalgleish R, Maglott DR, Hart RK, Greenblatt MS,
McGowan-​Jordan J, Roux AF, Smith T, Antonarakis SE, Taschner PE. HGVS
Recommendations for the Description of Sequence Variants: 2016 update.
Hum Mutat 2016, 37(6):564–​569.
101. Braschi B, Denny P, Gray K, Jones T, Seal R, Tweedie S, Yates B, Bruford E.
Genenames.org: the HGNC and VGNC resources in 2019. Nucleic Acids Res
2019, 47(D1):D786–​D792.
102. Arteche-​Lopez A, Avila-​Fernandez A, Romero R, Riveiro-​Alvarez R, Lopez-​
Martinez MA, Gimenez-​ Pardo A, Velez-​ Monsalve C, Gallego-​ Merlo J,
Garcia-​Vara I, Almoguera B et al. Sanger sequencing is no longer always
necessary based on a single-​center validation of 1109 NGS variants in 825
clinical exomes. Sci Rep 2021, 11(1):5697.
270 Next-Generation Sequencing Data Analysis

103. Beck TF, Mullikin JC, Program NCS, Biesecker LG. Systematic evaluation of
sanger validation of next-​generation sequencing variants. Clin Chem 2016,
62(4):647–​654.
104. Kerkhof J, Schenkel LC, Reilly J, McRobbie S, Aref-​Eshghi E, Stuart A, Rupar
CA, Adams P, Hegele RA, Lin H et al. Clinical validation of copy number
variant detection from targeted next-​generation sequencing panels. J Mol
Diagn 2017, 19(6):905–​920.
105. Miller DT, Lee K, Chung WK, Gordon AS, Herman GE, Klein TE, Stewart
DR, Amendola LM, Adelman K, Bale SJ et al. ACMG SF v3.0 list for reporting
of secondary findings in clinical exome and genome sequencing: a policy
statement of the American College of Medical Genetics and Genomics
(ACMG). Genet Med 2021, 23(8):1381–​1390.
106. Miller DT, Lee K, Gordon AS, Amendola LM, Adelman K, Bale SJ, Chung
WK, Gollob MH, Harrison SM, Herman GE et al. Recommendations for
reporting of secondary findings in clinical exome and genome sequencing,
2021 update: a policy statement of the American College of Medical Genetics
and Genomics (ACMG). Genet Med 2021, 23(8):1391–​1398.
107. Appelbaum PS, Parens E, Berger SM, Chung WK, Burke W. Is there a
duty to reinterpret genetic data? The ethical dimensions. Genet Med 2020,
22(3):633–​639.
108. Clayton EW, Appelbaum PS, Chung WK, Marchant GE, Roberts JL, Evans BJ.
Does the law require reinterpretation and return of revised genomic results?
Genet Med 2021, 23(5):833–​836.
109. Deignan JL, Chung WK, Kearney HM, Monaghan KG, Rehder CW, Chao
EC, Committee ALQA. Points to consider in the reevaluation and reanalysis
of genomic test results: a statement of the American College of Medical
Genetics and Genomics (ACMG). Genet Med 2019, 21(6):1267–​1270.
110. Roy S, Coldren C, Karunamurthy A, Kip NS, Klee EW, Lincoln SE, Leon
A, Pullambhatla M, Temple-​Smolkin RL, Voelkerding KV et al. Standards
and Guidelines for Validating Next-​Generation Sequencing Bioinformatics
Pipelines: A Joint Recommendation of the Association for Molecular
Pathology and the College of American Pathologists. J Mol Diagn 2018,
20(1):4–​27.
111. Ewing AD, Houlahan KE, Hu Y, Ellrott K, Caloian C, Yamaguchi TN, Bare JC,
P’ng C, Waggott D, Sabelnykova VY et al. Combining tumor genome simu-
lation with crowdsourcing to benchmark somatic single-​nucleotide-​variant
detection. Nat Methods 2015, 12(7):623–​630.
12 
De Novo Genome Assembly with
Long and/​or Short Reads

De novo genome assembly is a fundamental endeavor for genomics research,


as it creates the reference genome against which most other NGS applications
rely on for alignment. Accurate representation of the reference genome is of
utmost importance, as the assembly quality directly impacts the results of
other NGS applications that depend on it. Not very long ago Sanger sequen-
cing was considered the golden standard for de novo genome assembly.
However, it is prohibitively expensive and time-​ consuming to assemble
a genome using this first-​generation technology, as it took $3 billion and
13 years to generate the human genome draft assembly. The demand for
low-​cost and fast genome sequencing provides the very impetus for the
development of NGS technologies. The dramatically reduced cost of NGS
makes whole-​genome sequencing much more affordable, much faster, and
readily accessible to individual labs for deciphering any species. While
increasingly more and more organisms’ reference genomes are assembled,
de novo assembly continues to be an important application of NGS for not
only novel unsequenced genomes, but also completed genomes in terms of
resolving challenging regions (such as highly repetitive regions), removing
biases, and searching for novel components or structural variations. De novo
genome assembly from the relatively short and enormous number of reads
generated from most NGS platforms, however, poses serious challenges to
assembling algorithms that were designed for Sanger sequences. The usu-
ally shorter length of NGS reads means that they carry less information and
as a result lead to more uncertainties in the assembling process. To remedy
this situation, higher coverage is required, which significantly increases
the number of reads required and therefore computational complexity. For
example, using Sanger sequences with length up to 800 bp, assembling the
human genome used approximately 8× coverage; for NGS reads of 35–​100 bp,
the same task needs 50–​100× coverage [1].
Since Sanger sequence assemblers cannot effectively deal with these
challenges, new de novo genome assemblers have been developed for NGS
data. The development of Velvet [2] and ABySS [3] in 2008–​2009 showed
that de novo high-​quality genome assembly can be achieved, even for large
genomes, using massive numbers of ultra-​short (as short as 30 bp) reads.
The first de novo assembly of a human genome with the use of only short

DOI: 10.1201/9780429329180-15 271


272 Next-Generation Sequencing Data Analysis

NGS reads was accomplished in 2010 with the development SOAPdenovo


[4]. Subsequently, to increase assembly contiguity, a hybrid strategy is often
used to integrate long but error-​prone sequencing reads from PacBio (or
Nanopore) with short reads [5]. Now with significantly improved accuracy
of long-read sequencing, assembling a genome from long-read sequencing
data alone has become a reality [6].

12.1 Genomic Factors and Sequencing Strategies for


De Novo Assembly
12.1.1 Genomic Factors That Affect De Novo Assembly
The size of a target genome to a large degree determines the difficulty of
assembling it. All NGS de novo assemblers (to be detailed next) can handle
small genomes (<10 Mb), such as those of bacteria, without difficulty. For
genomes of medium size (10 Mb –​1 Gb), such as those of lower plants and
insects, most of the assemblers should still work without much problem.
For large genomes (>1 Gb), while some assemblers, such as the aforemen-
tioned SOAPdenovo, have been shown to have the capability to assemble the
human or other mammalian genomes, in general it is not an easy task to put
them together with only short reads. In addition, assembling a large genome
de novo is the most computationally demanding among all NGS applications.
The amount of repetitive sequences in a genome is another major factor
that affects de novo genome assembly. Some genomes are inherently more dif-
ficult to assemble than others because they contain more repetitive sequences.
Because they produce reads that are not unique due to their repetitive nature,
repetitive regions create serious challenges in the genome assembly pro-
cess. The challenges come from the inability to assemble reads from these
regions into contiguous segments (contigs) or scaffolds, and determine the
locations of these reads in relation to contigs or scaffolds assembled from
reads from non-​repetitive regions. As a result, these regions become gaps
in a draft assembly. Besides repetitive elements, genomic heterozygosity is
another factor that may affect de novo assembly. Genomic heterozygosity is a
measure of allelic differences in a genome, and allelic differences in a diploid
or polyploid genome lead to uncertainty in assembling their reads together.
In addition, other genome features, such as local GC content, may also affect
de novo genome assembly.

12.1.2 Sequencing Strategies for De Novo Assembly


Filling the gaps caused by repetitive regions is important for most de
novo genome assembly projects, and how to fill them should be a major
De Novo Genome Assembly with Long and/​or Short Reads 273

consideration when devising an appropriate sequencing strategy. The basic


approach to connect contigs or scaffolds across the gaps is to use read pairs,
or long reads, that span a distance longer than the gaps. For paired reads they
have to be generated from paired-​end and/​or mate-​pair sequencing, and
the known distances between the read pairs provide guidance to align the
contigs or scaffolds over the gaps. Mate-​pair sequencing differs from paired-​
end sequencing (see Chapter 4) in that the mate-​pair approach is designed to
“jump” sequence two ends of a larger DNA fragment. To conduct mate-​pair
sequencing, a DNA fragment is first circularized to have the two ends joined.
This circular DNA is then fragmented, and the segment that contains the
junction of the two ends is selected and sequenced with paired-​end sequen-
cing. To span repetitive regions in different sizes, sequencing reads generated
from mate-​pair libraries of varying insert sizes, e.g., from 2 to 40 Kb, as well
as regular paired-​end reads, are often used [7, 8].
The combined use of paired-​end and mate-​pair libraries of different insert
sizes is a key strategy in assembling a genome from NGS reads. The paired-​
end sequencing generates reads at the shorter size range (e.g., 300–​350 bp)
for assembling of non-​repeat sequences as well as resolving short repeat
sequences, while the mate-​pair “jump” sequencing produces reads at the
larger size range for resolving intermediate and long range repeat regions
and fill the corresponding gaps. Gaps of substantial sizes that are beyond the
covering range of mate pair libraries cannot be filled.
Besides the use of paired-​end and mate-​pair sequencing, read length is also
a key parameter for de novo genome assembly. While mammalian genomes
have been assembled from reads shorter than 75 bp (e.g., [4, 8]), longer reads
are always better. With long-read sequencing platforms such as PacBio
and ONT, obtaining long reads is becoming more routine. As detailed in
Chapter 4, CCS long reads from PacBio can reach 25 Kb, and 200 Kb can be
reached with CLR sequencing. On ONT flow cells, maximum read length
can reach over 4 Mb. In addition to native long reads, synthetic long reads
produced from approaches such as single-​tube Long Fragment Read (or
stLFR) from MGI [9] and Transposase Enzyme Linked Long-​read Sequencing
(TELL-​seq) from Universal Sequencing Technology [10] can reach 20–​300 Kb
and over 100 Kb, respectively. While synthetic long reads are based on short-
read sequencing, they differ from regular short reads in that instead of dir-
ectly breaking down genomic DNA into fragments of hundreds of base pairs
for sequencing, these approaches first shear DNA into rather large fragments
(up to hundreds of Kb). These large fragments are then used to produce short
fragments in such a way that short fragments derived from the same large
fragment all carry the same barcode, based on which short read sequences
are then linked together for synthesis of the original large fragment sequence.
10× Genomics introduced one of the first synthetic long-read strategies called
linked-​reads sequencing [11], but it was discontinued in 2020. Synthetic long
read approaches have greatly aided de novo genome assembly.
274 Next-Generation Sequencing Data Analysis

Sequencing depth is another important factor to consider for a de novo


assembly project. While it varies by project and is dependent on the other
factors (including the number of repeats and level of heterozygosity in the
genome as well as read length and error rate), a coverage that is too low
will undoubtedly result in a highly fragmented assembly. As a rough guide,
in the combined use of paired-​end and mate-​pair libraries of various insert
sizes, 45–​50x coverage is needed for the short-​insert-​size paired-​end and
intermediate-​size (3–​10 Kb) mate-​pair libraries, and 1–​5x coverage for the
long-​insert (10–​40 Kb) mate-​pair libraries [12, 13]. For de novo assembly of
a human genome using long reads alone, 30x or 35x coverage can be suffi-
cient for PacBio CCS or ONT reads respectively with appropriately chosen
tools [14, 15]. It should also be noted that while higher coverage may lead to
improvement in the final assembly quality, additional increase in coverage
also means increased data volume, computational complexity, and processing
time. There are also studies showing that beyond certain level of coverage
further increase in sequencing depth does not necessarily lead to increase in
assembly quality in terms of the size of assembled contigs [4].

12.2 Assembly of Contigs
12.2.1 Sequence Data Preprocessing, Error Correction,
and Assessment of Genome Characteristics
The de novo assembly of a genome from NGS reads is a multi-​step process
(Figure 12.1). As the first step, sequence data quality needs to be inspected.
Data QC steps described in Chapter 5 can be performed here to examine per-​
base error rate, quality score distribution, read size distribution, contamin-
ation of adaptor sequences, etc. Low-​quality reads need to be filtered out,
and portions of reads that contain low-​quality basecalls (usually the 3’ end),
ambiguities (reported as Ns), or adaptor sequences should be trimmed off.
As part of data preprocessing, paired-​end reads with part of their sequences
overlapped need to be merged to generate longer reads. The read merging
can also correct errors if discrepancy at some base positions are observed, in
which case the higher quality basecall is used. The merging process can be
handled by tools such as FLASH2 [16], PEAR [17], fastq-​join [18], PANDAseq
[19], and VSEARCH [20].
Sequencing error correction is an important step for de novo read assembly,
more so than for most other NGS applications due to the fact that the assembly
process is much more sensitive to these errors. The data QC measures
mentioned above cannot totally remove sequencing errors, as high basecall
quality scores alone cannot guarantee a read is free of sequencing errors.
If left uncorrected, the errors will lead to prolonged computational time,
De Novo Genome Assembly with Long and/​or Short Reads 275

Sequencing Paired-End &


Mate-Pair Libraries
(Of varying insert sizes)

Data QC & Preprocessing


(Incl. error correction &
genome size and coverage estimation)

Contig Assembly
(Greedy, OLC, & de Bruijn approaches)

Scaffold Construction

Genome Assembly
Quality Evaluation

Gap Closure

FIGURE 12.1
General workflow for de novo genome assembly.

erroneous contigs, and low-​quality genome assembly. While it can be time


consuming, an additional error correction step can improve final assembly
quality. There are multiple options to carry out this step. For example, BFC
[21], BLESS [22], Lighter [23], Musket [24], Fiona [25], and Coral [26] can
be used as stand-alone tools, while some assemblers (see next) have their
own error correction modules, such as ALLPATHS-​LG [27] and SGA [28].
Most error correction methods, including BFC, BLESS, Lighter, Musket, the
ALLPATHS-​LG error correction module (can be used stand-alone), and the
SGA default correction method, are based on k-​mer filtering [29]. K-​mer
refers to all the possible subsequences of length k in a read, and breaking
reads to k-​mers makes the complicated task of genome assembly more tract-
able. When all reads are converted to k-​mers, most k-​mers in the pool are
represented multiple times. Having a k-​mer that appears only once or twice
is an indication of sequencing error (Figure 12.2). The general error correction
approach is to find the smallest number of base changes to make all k-​mers
contained in a read “strong,” i.e., with the frequency of these k-​mers from all
reads above a threshold level. To determine the appropriate threshold level
for error correction, the distribution of the frequency of k-​mers can be plotted
using data from a k-​mer counting software such as Jellyfish [30] or Meryl [31].
Besides error correction, the k-​mer frequency information from Jellyfish or
276 Next-Generation Sequencing Data Analysis

0.015

Error k-mers
0.010

True k-mers
Density
0.005
0.000

0 20 40 60 80 100
Coverage
FIGURE 12.2
The coverage profile of true k-​mers and those with sequencing errors. (Adapted from Kelley
D.R. et al. (2010) Quake: quality-​aware detection and correction of sequencing errors, Genome
Biology, 11:R116. Used under the terms of the Creative Commons Attribution License (http://​
crea​tive​comm​ons.org/​licen​ses/​by/​2.0). © 2010 Kelley et al.)

Meryl can also be input into GenomeScope [32] to estimate genome size, level
of duplication, and heterozygosity. Besides using k-​mer filtering, suffix tree/​
array and multiple sequence alignment (MSA) are also often used to correct
sequencing errors. For example, Fiona is an example of the suffix tree/​array-​
based approach. Coral, on the other hand, is based on the MSA approach. The
approach aligns reads that share common k-​mers, and error corrections are
made based on alignment results and consensus sequences.
For long reads from PacBio or ONT that have higher error rates, sequencing
error correction is even more needed and can be performed with tools such
as FMLRC [33], PBcR [34], LoRDEC [35], LSC [36], Nanocorr [37], proovread
[38], and DeepConsensus [39]. Many long-read assemblers (to be detailed
next) contain their own error correction modules, such as Canu [40], Hifiasm
[41], FALCON [42], MARVEL [43], MECAT [44], and NECAT [14]. In general,
long-read error correction methods can be divided into two groups, with one
using redundant information in long reads alone for error correction, while the
other using a hybrid approach to leverage more accurate short reads to help
correct errors in long reads. Long-read-only correction methods belonging
to the first group include LoRMA [45] and FLAS [46]. The error correction
De Novo Genome Assembly with Long and/​or Short Reads 277

methods used by Canu, MARVEL, and MECAT, are also in this group since
they are based on consensus correction using sampling redundancy (or
overlaps) within long reads. As an example, to make corrections Canu builds
overlaps between long reads using a probabilistic sequence overlapping
algorithm called the MinHash alignment process (or MHAP) [47]. The over-
lapping results are then used to identify regions that need correction based
on sequence consensus, followed by estimation of corrected read length and
generation of corrected reads. Besides error correction, MARVEL also has a
“patching” module, which aims to repair apparent large-scale errors (e.g.,
regions that contain a lot of errors) based on comparisons between reads.
Examples of the hybrid approach to use more accurate short reads to correct
error-​prone long reads include FMLRC, PBcR, LoRDEC, LSC, Nanocorr, and
proovread. Based on how they work, these methods can be further divided
into those based on assembly and those on alignment. Assembly-​ based
correction methods, such as FMLRC and LoRDEC, first use short reads to
perform assembly, and then align long reads to the assemblies for making
corrections. Alignment-​based methods, including PBcR, LSC, Nanocorr, and
proovread, align short reads to long reads and sequencing errors in the long
reads are corrected based on the alignment results.

12.2.2 Contig Assembly Algorithms


Fundamentally different from the reference-​based alignment process, which
is used by most of the other NGS applications in this book, de novo genome
assembly attempts to construct a superstring (or superstrings) of DNA letters
based on the overlapping of sequence reads. This assembly process was pre-
viously modeled by Lander and Waterman with the use of ideal (error-​and
repeat-​free) sequence data [48]. In this model, if two reads overlap and the
overlap is above a cutoff level, the two reads are merged into a contig and
this process reiterates until the contig cannot be extended further. Although
this guiding model is straightforward, finding all possible overlaps between
millions of short reads and assembling them into contigs are computation-
ally intensive and challenging. Added to this challenge are other compli-
cating factors such as sequencing errors, sequencing bias, heterozygosity, and
repetitive sequences. To deal with these challenges, a number of assemblers
that employ different methodologies have been developed.
The currently available de novo genome assemblers can be classified into
three major categories –​those using (1) the Greedy approach; (2) the Overlap-​
Layout-​Consensus (OLC) approach; and (3) the de Bruijn graph. Although
all of them are based on graphs, the Greedy approach is the one that is based
on the maximization of local sequence similarity. It was used by Sanger
sequence assemblers, such as phrap and the TIGR assembler, and early NGS
reads assemblers, such as SSAKE [49], SHARCGS [50], and VCAKE [51].
Since it is a local approach, the Greedy approach does not consider the global
relationship between reads. Therefore, more recent NGS-​based assemblers
278 Next-Generation Sequencing Data Analysis

A B

R1 R2 R3 R4 R5 R6 K1 K2 K3 K14 K15 K16

FIGURE 12.3
Comparison of the OLC (A) and the de Bruijn graph (B) approaches for global de novo genome
assembly. In the OLC example, six sequence reads (R1–​R6) are shown for the illustrated genomic
region, with each read being 10 bp in length and the overlap between them set at ≥ 5 bp. The
reads are laid out in order based on how they overlap. The OLC graph is shown at the bottom,
with many nodes having more than one incoming or outgoing connections. In the de Bruijn
graph example, the reads are cut into a series of k-​mers (k=​5). In total there are 16 such k-​mers,
many of which occur in more than one read. The k-​mers are arranged sequentially based on how
they overlap, and the de Bruijn graph built from this approach is shown at the bottom. Different
from those in the OLC graph, the majority of the nodes in this graph have only one incoming
and one outgoing connection. (From Li Z. et al. Comparison of the two major classes of assembly
algorithms: overlap–​layout–​consensus and de-​bruijn-​graph. Briefings in Functional Genomics,
2012, 11(1): 25–​37, by permission of Oxford University Press.)

no longer use this approach, as it cannot take advantage of the global rela-
tionship offered by paired-​end and mate-​pair reads.
The OLC and the de Bruijn graph approaches are global by design and both
assemble reads into contigs using reads overlapping information based on
the Lander-​Waterman model. They approach the task, however, in different
ways (Figure 12.3). The OLC approach, as the name suggests, involves
three steps: (1) detecting potential overlaps between all reads; (2) laying
out all reads with their overlaps in a graph; and (3) constructing consensus
sequence superstring. The first step is computationally intensive and the
run time increases quadratically with the increase in the total number of
reads. The graph created in the second step consists of vertices (or nodes)
representing reads, and edges between them representing their overlaps.
The construction of a consensus sequence superstring equals to finding a
path in the graph that visits each node exactly once, which is known as a
Hamiltonian path in graph theory. While there are a small number of OLC-​
based short-read assemblers available such as Edena [52] and Fermi [53],
the OLC approach is more suitable and mostly used to assemble long reads
generated from PacBio and ONT sequencers. In fact, most long read de novo
assembly methods are based on OLC, including Canu, FALCON, Hifiasm,
De Novo Genome Assembly with Long and/​or Short Reads 279

MECAT, NECAT, miniasm [54], HINGE [55], Peregrine [56], Shasta [57],
Raven [58], NextDenovo [59], and SMARTdenovo [60]. In the case of Canu,
after the aforementioned error correction process, unsupported sequences
are trimmed off to prepare corrected reads for assembly. In the assembly
stage, reads are scanned one last time for errors, and then used to construct
overlap graph, before output of consensus contig sequences and an assembly
graph. HiCanu is a modified version of Canu developed for using the PacBio
HiFi (CCS) reads [61].
Solving the Hamiltonian path problem in the OLC approach is NP-​hard. To
reduce the high computing demand imposed by the OLC approach, a simpli-
fied version called the String graph has been employed to merge and reduce
redundant vertices and edges, along with identification and removal of false
vertices and edges [62]. The implementation of a string indexing data struc-
ture (FM-​index) has improved the performance of assemblers such as SGA
and ReadJoiner [63]. FALCON also first builds a string graph that contains
bubbles representing structural variation between haplotypes through the
use of a process called HGAP (or hierarchical genome assembly process). The
subsequent “Unzip” process creates haplotype-​ resolved assembly graph,
making FALCON a diploid-​aware assembler. Other long-read assemblers,
such as Miniasm, Hifiasm, NECAT, and NextDenovo, also use string graph
in their assembly processes. Hifiasm, for example, produces fully phased
assembly for each haplotype from phased string graph created from PacBio
HiFi reads.
Compared to the OLC approach, the de Bruijn graph-​based approach takes
an alternative, computationally more tractable route. This approach does not
involve a step to find all possible overlaps between reads. Instead, the reads
are first cut into k-​mers. For instance, the sequence read ATTACGTCGA can
be cut into a series of k-​mers, e.g., ATT, TTA, TAC, ACG, CGT, GTC, TCG,
and CGA, when k =​3. These k-​mers are then used as vertices in the de Bruijn
graph. An edge that connects two nodes represents a convergence of the
two nodes, e.g., the edge that connects ATT and TTA is ATTA. Using the de
Bruijn graph, the assembly process is equivalent to finding a shortest path
that visits each node at least once, which is known as the Chinese Postman
Problem in graph theory. An Eulerian path, if it exists, represents the solu-
tion to this problem. Computationally, finding an Eulerian path is much
easier than finding a Hamiltonian path for the OLC approach. The major
drawback of this approach, however, is that it is highly sensitive to sequen-
cing errors. Therefore, to use assemblers in this category, error correction
is mandatory. Assemblers that use this approach include AbySS [3],
ALLPATHS-​LG, Euler-​SR [29], SOAPdenovo/​SOAPdenovo2 [4, 64], SPAdes
[65], and Velvet [2]. Because of higher sequencing error rates, long reads
are not usually assembled with this approach. There are, however, a small
number of long read assemblers that employ variants of de Bruijn Graph.
For example, Flye [66] uses the A-​Bruijn variant, which tolerates the higher
280 Next-Generation Sequencing Data Analysis

level of sequencing errors in long reads as it uses approximate sequence


matches instead of exact k-​mer matches. Using PacBio or ONT long reads as
input, it first creates a repeat graph, and then through resolving repetitions
by identifying variations in repeat copies in the graph a final assembly is
reconstructed. Wtdbg2 [67] is another example based on another de Bruijn
variant called fuzzy Bruijn graph (or FBG), which permits mismatches and
gaps. This method assembles a genome through first cutting long reads into
smaller segments, and then merging the segments based on their adjacency
to create contigs.
Hybrid assemblers integrate both short and long reads toward achieving
improved accuracy and contiguity. Some of the assemblers, such as MaSuRCA
[68] and DBG2OLC [69], use a combination of the de Bruijn graph and OLC
approaches for short and long reads, respectively. Others take alternative
approaches. For example, WENGAN uses de Bruijn graph to assemble short
reads, but avoids the all-​versus-​all read comparison used in the OLC process
commonly used for long reads to increase efficiency [70].

12.2.3 Polishing
Besides error correction prior to genome assembly, assembly quality can be
further improved after draft assembly is created through a process called
polishing. In general terms, the polishing process uses information from
alignment of reads to the draft assembly as input, examines how reads map
to the draft assembly at each location, and then decides whether assembly
sequences at certain locations need to be modified. To perform this process,
there are a selection of tools available, including Pilon [71], Medaka [72],
Racon [73], Nanopolish [74], MarginPolish & HELEN [57], NextPolish [75],
POLCA [76], and NeuralPolish [77]. These polishers typically use either
short or long reads to polish a draft assembly. For example, Pilon takes as
input an assembled genome in FASTA format, and alignment of short reads
to the assembly in BAM format. After searching the alignment for incon-
sistencies, assembly sequences are modified to provide improvements to
the input assembly through reduction of mismatches and indels, as well
as gap filling and misassembly identification. Medaka, the first neural
network-​ based polisher, on the other hand, uses ONT long reads for
polishing through creation of consensus sequences. As input it requires a
draft assembly in FASTA format and basecalls in either FASTA or FASTQ
format. Prior to creating consensus sequences, the alignment of the reads
to the input assembly is carried out by minimap2. Nanopolish uses a third
approach. Instead of using called bases, it takes raw sequencing signals
recorded from an ONT sequencer as input, and applies an HMM-​based
signal-​to-​assembly analysis to generate improved consensus sequences for
the draft assembly.
De Novo Genome Assembly with Long and/​or Short Reads 281

12.3 Scaffolding and Gap Closure


After assembly of contigs, the next step is to organize the contigs into a
“scaffold” structure to improve continuity rather than leave them disjointed.
This scaffolding process orders and orients the contigs, and estimates the
lengths of the gaps between them (Figure 12.4). To establish positional rela-
tionship between contigs, scaffolding algorithms use long reads or mate-​pair
reads that span different contigs.
For input, scaffolding algorithms typically take pre-​assembled contigs, and
long reads or sometimes mate-​pair reads. The first and also an important
step in the scaffolding process is to map the input long reads or read pairs
to the contigs. To improve mapping results, sequencing errors in the reads
should be corrected prior to mapping as covered above. To assemble the
contigs into scaffolds using the guiding information in the long or mate-​pair
reads, scaffolders usually take a graph-​based approach similar to the contig
assembly process, but here with contigs as nodes and connecting long reads
(or read pairs) as edges. The quality of the assembled scaffolds is dependent
on the quality of input contigs, the complexity of the genome, and the quality
of long or mate-​pair read libraries. The sizes of the scaffolds are limited
by the length of long reads or the insert size of mate-​pair libraries, as the
scaffolds cannot span repetitive regions larger than the insert size or read
length. Besides long reads or mate-​pair reads, scaffolding information may
also come from experimental data generated from other techniques including
Hi-​C, synthetic long-read sequencing, and optical mapping.
Currently available stand-alone scaffolders include SSPACE-​LongRead [78],
LINKS [79], OPERA-​LG [80], LRScaf [81], SMIS [82], and npGraph [83]. To

Scaffold

Contig 1 Contig 2

DNA Fragments
Paired-end read
Approximate length, but no known sequence
FIGURE 12.4
Assembling contigs into a scaffold.
282 Next-Generation Sequencing Data Analysis

provide an example of how scaffolding can be performed, LRScaf first maps


long reads using minimap2 to identify those that overlap the ends of contigs,
and then connects contigs based on reads that map to multiple contigs. From
the connections, a scaffold graph similar to a string graph is constructed, and
scaffolding of contigs is achieved through traversal of the graph to expose
all linear paths. Besides these stand-alone methods, many contig assemblers,
including ABySS, SGA, SOAPdenovo2, SPAdes, and MaSuRCA, DBG2OLC,
also have built-​ in scaffolding modules. The performance of different
scaffolders varies with data sets and analysis parameters. Therefore, before
deciding on an appropriate scaffolder for a project, it is helpful to first try out
different scaffolders using different parameters and then evaluate the results
(see Section 12.4).
Among the other techniques that generate data for scaffolding, Hi-​C is an
NGS-​based approach that uses proximity ligation to interrogate chromo-
somal interactions within the nucleus. The principle behind the use of Hi-​
C for scaffolding is that the probability of intrachromosomal interactions is
much higher than that of interchromosomal contacts. Therefore, Hi-​C reads
provide long-​range information on linear arrangement of contig sequences
along individual chromosomes. Scaffolders that utilize Hi-​C data include
HiRise [84], 3D-​DNA [85], and SALSA [86]. Similar to native long reads
generated from ONT and PacBio sequencers, synthetic long reads also pro-
vide long-​range sequence information required for scaffolding of contigs.
Currently available scaffolding methods that use synthetic long reads include
ARCS [87], Architect [88], ARKS [87], and SLR-​superscaffolder [89].
The final stage of finishing a de novo genome assembly is to close the
gaps between scaffolded contigs. Gap closure has greatly benefited from
the constant advancements in long reads sequencing. There are currently a
number of gap filling algorithms available, including PBjelly [5], GMcloser
[90], Sealer [91], LR_​Gapcloser [92], TGS-​GapCloser [93], SAMBA [94], and
gapless [95]. Among these algorithms SAMBA and gapless can perform both
scaffolding and gap filling. Some assemblers, such as ABySS, ALLPATHS-​
LG, and SOAPdenovo2, also contain gap filling modules. Figure 12.5 shows
an example of how long reads are used to finish gaps by TGS-​GapCloser.

12.4 Assembly Quality Evaluation


Contiguity, completeness, and accuracy are key indices of the quality of
an assembly. Contiguity is reflected by the total number of assembled
contigs or scaffolds and their size distribution, i.e., whether the assembly
is composed of a small number of large fragments or a large number of
small fragments. It can be measured by statistics such as mean or median
De Novo Genome Assembly with Long and/​or Short Reads 283

FIGURE 12.5
Gap closing with the TGS-​GapCloser workflow. Panel (A) displays the overall pipeline. Panel
(B) shows the steps of identification of gap regions and long-​read candidates that map to the
gap regions, and error correction of long-​read candidates using short reads. Panel (C) details
how the gaps are filled with error-​corrected long reads. As input, TGS-​GapCloser can accept
long reads generated from any platform or other preassembled contigs to fill gaps in a draft
assembly. The unknown nucleotides marked as N’s between two neighboring contigs in the
scaffolds are identified as gap regions. Long reads are then mapped to the gap regions using
minimap2 to acquire candidate fragments. The subsequent error correction on the long-​read
candidates is carried out with Pilon or Racon. The corrected new long read candidates are then
realigned to the gaps, and the read with the best match for a gap is used to fill the gap. To increase
computational efficiency, overlapped candidates are clipped and merged. (Adapted from Xu
M. et al. (2020) TGS-​GapCloser: A fast and accurate gap closer for large genomes with low
coverage of error-​prone long reads. GigaScience, 9, 1–​11. Used under the terms of the Creative
Commons Attribution License (http://​crea​tive​comm​ons.org/​licen​ses/​by/​4.0). © 2020 Xu et al.)
284 Next-Generation Sequencing Data Analysis

length, but the most commonly used statistics are N50 and L50. To calcu-
late the N50, all contigs (or scaffolds) are first ranked based on length from
the largest to the smallest. Their lengths are then summed up from the
largest contig (or scaffold) downward. N50 refers to the size of the contig
(or scaffold) at which the summed length becomes greater or equal to 50%
of the total assembly size. L50, on the other hand, refers to the smallest
number of contigs (or scaffolds) that add up to at least 50% of the genome
assembly.
The total assembly size, however, does not measure the completeness of
the assembly. To determine completeness, the original DNA reads are aligned
to the assembly and the percentage of reads aligned is calculated. Other
sequence data from the same species, such as RNA-​seq data, may also be used
for the alignment and rough estimation of completeness. On the measure-
ment of accuracy, the assembly can be compared to a high-​quality reference
genome of the species, if such a reference is available. This comparison can
be carried out on two aspects of the assembly: base accuracy and alignment
accuracy. Base accuracy determines if the right base is called in the assembly
at a given position, while alignment accuracy examines the probability of
placing a sequence at the right position and orientation. In many cases,
however, a reference map is not available and instead is the very goal of the
assembling process. For these cases, a measurement on internal consistency,
through aligning original reads to the assembly and checking for evenness
and congruence in coverage across the assembly, provides an indicator of the
assembly quality. Comparison of the assembly with independently acquired
sequences from the same species, such as gene or cDNA sequences, can also
be used to estimate assembly accuracy. With regard to software implementa-
tion on evaluating assembly quality, the often used tools include BUSCO [96]
and QUAST [97], which help perform the above measurements and compare
different contig and scaffold assembly algorithms and settings.

12.5 Limitations and Future Development


Among all NGS applications covered in this book, de novo genome assembly
remains one of the most challenging due to limitations of current sequen-
cing technologies and the complexity of genomic landscape. Due to their
cost effectiveness, short read sequencing remains to be the most accessible;
however, the short read length poses a limit on de novo genome assembly.
This, combined with other factors including sequencing errors, repetitive
elements, and uneven regional coverage, leads to ambiguities, false-​positive
and branched paths in the assembly graph, and early terminations in contig
extension, limiting the completeness of assembled sequences. As a result,
De Novo Genome Assembly with Long and/​or Short Reads 285

the assembled sequences are usually fragmented and exist in the suboptimal
form of large numbers of contigs. Among the contigs, there are also cer-
tain (sometimes high) levels of falsely assembled contigs, due to chimeric
joining. In addition, the gapped regions between the assembled contigs may
not be filled completely. To overcome some of these limitations and increase
assembly quality, the use of a reference genome, even from a remotely related
species, can be very helpful. This reference-​ assisted assembly approach
works especially well when scaffolding information from paired reads is not
available or exhausted. With the quickly increasing number of sequenced
genomes, improving assembly quality with this reference-​assisted approach
becomes more feasible. Some tools have recently been developed to pro-
vide this functionality, either as dedicated packages such as RaGOO [98],
Chromosomer [99], and RACA [100], or components of existing assemblers
including ALLPATHS-​LG, IDBA-​Hybrid, and Velvet.
With the constant advancements achieved in long read sequencing,
the landscape of de novo genome assembly has been shifting. The publica-
tion of a gapless human genome assembly by the Telomere-​to-​Telomere
(T2T) Consortium has demonstrated the utility of long reads in de novo
genome assembly [101]. Built on this success, more and better designed
assemblers are bound to be continuously developed to take full advantage
of what new sequencing technologies have to offer. Undoubtedly such future
developments will further overcome the limitations and challenges facing
the community today.

References
1. Schatz MC, Delcher AL, Salzberg SL. Assembly of large genomes using
second-​generation sequencing. Genome Res 2010, 20(9):1165–​1173.
2. Zerbino DR, Birney E. Velvet: algorithms for de novo short read assembly
using de Bruijn graphs. Genome Res 2008, 18(5):821–​829.
3. Simpson JT, Wong K, Jackman SD, Schein JE, Jones SJ, Birol I. ABySS: a parallel
assembler for short read sequence data. Genome Res 2009, 19(6):1117–​1123.
4. Li R, Zhu H, Ruan J, Qian W, Fang X, Shi Z, Li Y, Li S, Shan G, Kristiansen K
et al. De novo assembly of human genomes with massively parallel short read
sequencing. Genome Res 2010, 20(2):265–​272.
5. English AC, Richards S, Han Y, Wang M, Vee V, Qu J, Qin X, Muzny DM,
Reid JG, Worley KC et al. Mind the gap: upgrading genomes with Pacific
Biosciences RS long-​read sequencing technology. PLoS One 2012, 7(11):e47768.
6. Rayamajhi N, Cheng CC, Catchen JM. Evaluating Illumina-​, Nanopore-​, and
PacBio-​based genome assembly strategies with the bald notothen, Trematomus
borchgrevinki. G3 2022, 12(11):jkac192.
7. van Heesch S, Kloosterman WP, Lansu N, Ruzius FP, Levandowsky E, Lee CC,
Zhou S, Goldstein S, Schwartz DC, Harkins TT et al. Improving mammalian
286 Next-Generation Sequencing Data Analysis

genome scaffolding using large insert mate-​pair next-​generation sequencing.


BMC Genomics 2013, 14:257.
8. Li R, Fan W, Tian G, Zhu H, He L, Cai J, Huang Q, Cai Q, Li B, Bai Y et al.
The sequence and de novo assembly of the giant panda genome. Nature 2010,
463(7279):311–​317.
9. Wang O, Chin R, Cheng X, Wu MKY, Mao Q, Tang J, Sun Y, Anderson E, Lam
HK, Chen D et al. Efficient and unique cobarcoding of second-​generation
sequencing reads from long DNA molecules enabling cost-​ effective and
accurate sequencing, haplotyping, and de novo assembly. Genome Res 2019,
29(5):798–​808.
10. Chen Z, Pham L, Wu TC, Mo G, Xia Y, Chang PL, Porter D, Phan T, Che H,
Tran H et al. Ultralow-​input single-​tube linked-​read library method enables
short-​read second-​ generation sequencing systems to routinely generate
highly accurate and economical long-​range sequencing information. Genome
Res 2020, 30(6):898–​909.
11. Zheng GX, Lau BT, Schnall-​ Levin M, Jarosz M, Bell JM, Hindson CM,
Kyriazopoulou-​Panagiotopoulou S, Masquelier DA, Merrill L, Terry JM et al.
Haplotyping germline and cancer genomes with high-​throughput linked-​read
sequencing. Nat Biotechnol 2016, 34(3):303–​311.
12. Nagarajan N, Pop M. Sequence assembly demystified. Nature Rev Genet 2013,
14(3):157–​167.
13. Desai A, Marwah VS, Yadav A, Jha V, Dhaygude K, Bangar U, Kulkarni V, Jere
A. Identification of optimum sequencing depth especially for de novo genome
assembly of small genomes using next generation sequencing data. PLoS One
2013, 8(4):e60204.
14. Chen Y, Nie F, Xie SQ, Zheng YF, Dai Q, Bray T, Wang YX, Xing JF, Huang ZJ,
Wang DP et al. Efficient assembly of nanopore reads via highly accurate and
intact error correction. Nature Commun 2021, 12(1):60.
15. Chen Y, Zhang Y, Wang AY, Gao M, Chong Z. Accurate long-​read de novo
assembly evaluation with Inspector. Genome Biol 2021, 22(1):312.
16. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve
genome assemblies. Bioinformatics 2011, 27(21):2957–​2963.
17. Zhang J, Kobert K, Flouri T, Stamatakis A. PEAR: a fast and accurate Illumina
Paired-​End reAd mergeR. Bioinformatics 2014, 30(5):614–​620.
18. Aronesty E. Comparison of sequencing utility programs. Open Bioinformatics J
2013, 7(1):1–​8.
19. Masella AP, Bartram AK, Truszkowski JM, Brown DG, Neufeld JD.
PANDAseq: paired-​end assembler for Illumina sequences. BMC Bioinformatics
2012, 13:31.
20. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open
source tool for metagenomics. PeerJ 2016, 4:e2584.
21. Li H. BFC: correcting Illumina sequencing errors. Bioinformatics 2015,
31(17):2885–​2887.
22. Heo Y, Wu XL, Chen D, Ma J, Hwu WM. BLESS: bloom filter-​based error
correction solution for high-​throughput sequencing reads. Bioinformatics 2014,
30(10):1354–​1362.
23. Song L, Florea L, Langmead B. Lighter: fast and memory-​efficient sequencing
error correction without counting. Genome Biol 2014, 15(11):509.
De Novo Genome Assembly with Long and/​or Short Reads 287

24. Liu Y, Schroder J, Schmidt B. Musket: a multistage k-​mer spectrum-​based


error corrector for Illumina sequence data. Bioinformatics 2013, 29(3):308–​315.
25. Schulz MH, Weese D, Holtgrewe M, Dimitrova V, Niu S, Reinert K, Richard H.
Fiona: a parallel and automatic strategy for read error correction. Bioinformatics
2014, 30(17):i356–​363.
26. Salmela L, Schroder J. Correcting errors in short reads by multiple alignments.
Bioinformatics 2011, 27(11):1455–​1461.
27. Gnerre S, Maccallum I, Przybylski D, Ribeiro FJ, Burton JN, Walker BJ, Sharpe
T, Hall G, Shea TP, Sykes S et al. High-​quality draft assemblies of mammalian
genomes from massively parallel sequence data. Proc Natl Acad Sci U S A 2011,
108(4):1513–​1518.
28. Simpson JT, Durbin R. Efficient de novo assembly of large genomes using
compressed data structures. Genome Res 2012, 22(3):549–​556.
29. Pevzner PA, Tang H, Waterman MS. An Eulerian path approach to DNA
fragment assembly. Proc Natl Acad Sci U S A 2001, 98(17):9748–​9753.
30. Marcais G, Kingsford C. A fast, lock-​ free approach for efficient parallel
counting of occurrences of k-​mers. Bioinformatics 2011, 27(6):764–​770.
31. Rhie A, Walenz BP, Koren S, Phillippy AM. Merqury: reference-​free quality,
completeness, and phasing assessment for genome assemblies. Genome Biol
2020, 21(1):245.
32. Ranallo-​Benavidez TR, Jaron KS, Schatz MC. GenomeScope 2.0 and
Smudgeplot for reference-​free profiling of polyploid genomes. Nat Commun
2020, 11(1):1432.
33. Wang JR, Holt J, McMillan L, Jones CD. FMLRC: Hybrid long read error
correction using an FM-​index. BMC Bioinformatics 2018, 19(1):50.
34. Koren S, Schatz MC, Walenz BP, Martin J, Howard JT, Ganapathy G, Wang
Z, Rasko DA, McCombie WR, Jarvis ED et al. Hybrid error correction and
de novo assembly of single-​molecule sequencing reads. Nat Biotechnol 2012,
30(7):693–​700.
35. Salmela L, Rivals E. LoRDEC: accurate and efficient long read error correction.
Bioinformatics 2014, 30(24):3506–​3514.
36. Au KF, Underwood JG, Lee L, Wong WH. Improving PacBio long read
accuracy by short read alignment. PLoS One 2012, 7(10):e46679.
37. Goodwin S, Gurtowski J, Ethe-​Sayers S, Deshpande P, Schatz MC, McCombie
WR. Oxford Nanopore sequencing, hybrid error correction, and de novo
assembly of a eukaryotic genome. Genome Res 2015, 25(11):1750–​1756.
38. Hackl T, Hedrich R, Schultz J, Forster F. proovread: large-​scale high-​accuracy
PacBio correction through iterative short read consensus. Bioinformatics 2014,
30(21):3004–​3011.
39. Baid G, Cook DE, Shafin K, Yun T, Llinares-​López F, Berthet Q, Wenger AM,
Rowell WJ, Nattestad M, Yang H et al. DeepConsensus improves the accuracy
of sequences with a gap-​aware sequence transformer. Nat Biotechnol 2023,
41(2):232–​238.
40. Koren S, Walenz BP, Berlin K, Miller JR, Bergman NH, Phillippy AM.
Canu: scalable and accurate long-​read assembly via adaptive k-​mer weighting
and repeat separation. Genome Res 2017, 27(5):722–​736.
41. Cheng H, Concepcion GT, Feng X, Zhang H, Li H. Haplotype-​resolved de
novo assembly using phased assembly graphs with Hifiasm. Nat Methods 2021,
18(2):170–​175.
288 Next-Generation Sequencing Data Analysis

42. Chin CS, Peluso P, Sedlazeck FJ, Nattestad M, Concepcion GT, Clum A, Dunn
C, O’Malley R, Figueroa-​Balderas R, Morales-​Cruz A et al. Phased diploid
genome assembly with single-​molecule real-​time sequencing. Nat Methods
2016, 13(12):1050–​1054.
43. Nowoshilow S, Schloissnig S, Fei JF, Dahl A, Pang AWC, Pippel M, Winkler S,
Hastie AR, Young G, Roscito JG et al. The axolotl genome and the evolution of
key tissue formation regulators. Nature 2018, 554(7690):50–​55.
44. Xiao CL, Chen Y, Xie SQ, Chen KN, Wang Y, Han Y, Luo F, Xie Z. MECAT: fast
mapping, error correction, and de novo assembly for single-​molecule sequen-
cing reads. Nat Methods 2017, 14(11):1072–​1074.
45. Salmela L, Walve R, Rivals E, Ukkonen E. Accurate self-​correction of errors in
long reads using de Bruijn graphs. Bioinformatics 2017, 33(6):799–​806.
46. Bao E, Xie F, Song C, Song D. FLAS: fast and high-​throughput algorithm for
PacBio long-​read self-​correction. Bioinformatics 2019, 35(20):3953–​3960.
47. Berlin K, Koren S, Chin CS, Drake JP, Landolin JM, Phillippy AM. Assembling
large genomes with single-​ molecule sequencing and locality-​ sensitive
hashing. Nat Biotechnol 2015, 33(6):623–​630.
48. Lander ES, Waterman MS. Genomic mapping by fingerprinting random
clones: a mathematical analysis. Genomics 1988, 2(3):231–​239.
49. Warren RL, Sutton GG, Jones SJ, Holt RA. Assembling millions of short DNA
sequences using SSAKE. Bioinformatics 2007, 23(4):500–​501.
50. Dohm JC, Lottaz C, Borodina T, Himmelbauer H. SHARCGS, a fast and highly
accurate short-​read assembly algorithm for de novo genomic sequencing.
Genome Res 2007, 17(11):1697–​1706.
51. Jeck WR, Reinhardt JA, Baltrus DA, Hickenbotham MT, Magrini V, Mardis ER,
Dangl JL, Jones CD. Extending assembly of short DNA sequences to handle
error. Bioinformatics 2007, 23(21):2942–​2944.
52. Hernandez D, Francois P, Farinelli L, Osteras M, Schrenzel J. De novo bacterial
genome sequencing: millions of very short reads assembled on a desktop com-
puter. Genome Res 2008, 18(5):802–​809.
53. Li H. Exploring single-​sample SNP and INDEL calling with whole-​genome de
novo assembly. Bioinformatics 2012, 28(14):1838–​1844.
54. Li H. Minimap and miniasm: fast mapping and de novo assembly for noisy
long sequences. Bioinformatics 2016, 32(14):2103–​2110.
55. Kamath GM, Shomorony I, Xia F, Courtade TA, Tse DN. HINGE: long-​
read assembly achieves optimal repeat resolution. Genome Res 2017,
27(5):747–​756.
56. Chin C-​S, Khalak A. Human Genome Assembly in 100 Minutes. bioRxiv 2019,
doi: https://​doi.org/​10.1101/​705​616
57. Shafin K, Pesout T, Lorig-​Roach R, Haukness M, Olsen HE, Bosworth C,
Armstrong J, Tigyi K, Maurer N, Koren S et al. Nanopore sequencing and the
Shasta toolkit enable efficient de novo assembly of eleven human genomes.
Nat Biotechnol 2020, 38(9):1044–​1053.
58. Vaser R, Šikić M. Time-​and memory-​efficient genome assembly with Raven.
Nat Comput Sci 2021, 1(5):332–​336.
59. NextDenovo (https://​git​hub.com/​Nextom​ics/​Nex​tDen​ovo)
60. Liu H, Wu S, Li A, Ruan J. SMARTdenovo: a de novo assembler using long
noisy reads. Gigabyte 2021:1–​9.
De Novo Genome Assembly with Long and/​or Short Reads 289

61. Nurk S, Walenz BP, Rhie A, Vollger MR, Logsdon GA, Grothe R, Miga KH,
Eichler EE, Phillippy AM, Koren S. HiCanu: accurate assembly of segmental
duplications, satellites, and allelic variants from high-​ fidelity long reads.
Genome Res 2020, 30(9):1291–​1305.
62. Myers EW. Toward simplifying and accurately formulating fragment
assembly. J Comput Biol 1995, 2(2):275–​290.
63. Gonnella G, Kurtz S. Readjoiner: a fast and memory efficient string graph-​
based sequence assembler. BMC Bioinformatics 2012, 13:82.
64. Luo R, Liu B, Xie Y, Li Z, Huang W, Yuan J, He G, Chen Y, Pan Q, Liu Y et al.
SOAPdenovo2: an empirically improved memory-​efficient short-​read de novo
assembler. GigaScience 2012, 1(1):18.
65. Nurk S, Bankevich A, Antipov D, Gurevich AA, Korobeynikov A, Lapidus A,
Prjibelski AD, Pyshkin A, Sirotkin A, Sirotkin Y et al. Assembling single-​cell
genomes and mini-​metagenomes from chimeric MDA products. J Comput Biol
2013, 20(10):714–​737.
66. Kolmogorov M, Yuan J, Lin Y, Pevzner PA. Assembly of long, error-​prone
reads using repeat graphs. Nat Biotechnol 2019, 37(5):540–​546.
67. Ruan J, Li H. Fast and accurate long-​read assembly with wtdbg2. Nat Methods
2020, 17(2):155–​158.
68. Zimin AV, Marcais G, Puiu D, Roberts M, Salzberg SL, Yorke JA. The MaSuRCA
genome assembler. Bioinformatics 2013, 29(21):2669–​2677.
69. Ye C, Hill CM, Wu S, Ruan J, Ma ZS. DBG2OLC: Efficient Assembly of Large
Genomes Using Long Erroneous Reads of the Third Generation Sequencing
Technologies. Sci Rep 2016, 6:31900.
70. Di Genova A, Buena-​Atienza E, Ossowski S, Sagot MF. Efficient hybrid de
novo assembly of human genomes with WENGAN. Nat Biotechnol 2021,
39(4):422–​430.
71. Walker BJ, Abeel T, Shea T, Priest M, Abouelliel A, Sakthikumar S, Cuomo CA,
Zeng Q, Wortman J, Young SK et al. Pilon: an integrated tool for comprehen-
sive microbial variant detection and genome assembly improvement. PLoS
One 2014, 9(11):e112963.
72. Medaka (https://​git​hub.com/​nanop​oret​ech/​med​aka)
73. Vaser R, Sovic I, Nagarajan N, Sikic M. Fast and accurate de novo genome
assembly from long uncorrected reads. Genome Res 2017, 27(5):737–​746.
74. Loman NJ, Quick J, Simpson JT. A complete bacterial genome assembled
de novo using only nanopore sequencing data. Nat Methods 2015,
12(8):733–​735.
75. Hu J, Fan J, Sun Z, Liu S. NextPolish: a fast and efficient genome polishing tool
for long-​read assembly. Bioinformatics 2020, 36(7):2253–​2255.
76. Zimin AV, Salzberg SL. The genome polishing tool POLCA makes fast
and accurate corrections in genome assemblies. PLoS Comput Biol 2020,
16(6):e1007981.
77. Huang N, Nie F, Ni P, Luo F, Gao X, Wang J. NeuralPolish: a novel Nanopore
polishing method based on alignment matrix construction and orthogonal Bi-​
GRU Networks. Bioinformatics 2021, 37(19):3120–​3127.
78. Boetzer M, Pirovano W. SSPACE-​ LongRead: scaffolding bacterial draft
genomes using long read sequence information. BMC Bioinformatics 2014,
15:211.
290 Next-Generation Sequencing Data Analysis

79. Warren RL, Yang C, Vandervalk BP, Behsaz B, Lagman A, Jones SJ, Birol I.
LINKS: Scalable, alignment-​free scaffolding of draft genomes with long reads.
GigaScience 2015, 4:35.
80. Gao S, Bertrand D, Chia BK, Nagarajan N. OPERA-​LG: efficient and exact
scaffolding of large, repeat-​rich eukaryotic genomes with performance guar-
antees. Genome Biol 2016, 17:102.
81. Qin M, Wu S, Li A, Zhao F, Feng H, Ding L, Ruan J. LRScaf: improving draft
genomes using long noisy reads. BMC Genomics 2019, 20(1):955.
82. SMIS (Single Molecular Integrative Scaffolding) (www.san​ger.ac.uk/​tool/​
smis/​)
83. Nguyen SH, Cao MD, Coin LJM. Real-​time resolution of short-​read assembly
graph using ONT long reads. PLoS Comput Biol 2021, 17(1):e1008586.
84. Putnam NH, O’Connell BL, Stites JC, Rice BJ, Blanchette M, Calef R, Troll
CJ, Fields A, Hartley PD, Sugnet CW et al. Chromosome-​ scale shotgun
assembly using an in vitro method for long-​range linkage. Genome Res 2016,
26(3):342–​350.
85. Dudchenko O, Batra SS, Omer AD, Nyquist SK, Hoeger M, Durand NC,
Shamim MS, Machol I, Lander ES, Aiden AP et al. De novo assembly of the
Aedes aegypti genome using Hi-​ C yields chromosome-​ length scaffolds.
Science 2017, 356(6333):92–​95.
86. Ghurye J, Rhie A, Walenz BP, Schmitt A, Selvaraj S, Pop M, Phillippy AM,
Koren S. Integrating Hi-​C links with assembly graphs for chromosome-​scale
assembly. PLoS Comput Biol 2019, 15(8):e1007273.
87. Yeo S, Coombe L, Warren RL, Chu J, Birol I. ARCS: scaffolding genome drafts
with linked reads. Bioinformatics 2018, 34(5):725–​731.
88. Kuleshov V, Snyder MP, Batzoglou S. Genome assembly from synthetic long
read clouds. Bioinformatics 2016, 32(12):i216–​i224.
89. Guo L, Xu M, Wang W, Gu S, Zhao X, Chen F, Wang O, Xu X, Seim I, Fan G
et al. SLR-​superscaffolder: a de novo scaffolding tool for synthetic long reads
using a top-​to-​bottom scheme. BMC Bioinformatics 2021, 22(1):158.
90. Kosugi S, Hirakawa H, Tabata S. GMcloser: closing gaps in assemblies accur-
ately with a likelihood-​based selection of contig or long-​read alignments.
Bioinformatics 2015, 31(23):3733–​3741.
91. Paulino D, Warren RL, Vandervalk BP, Raymond A, Jackman SD, Birol I.
Sealer: a scalable gap-​closing application for finishing draft genomes. BMC
Bioinformatics 2015, 16:230.
92. Xu GC, Xu TJ, Zhu R, Zhang Y, Li SQ, Wang HW, Li JT. LR_​Gapcloser: a tiling
path-​based gap closer that uses long reads to complete genome assembly.
GigaScience 2019, 8(1):giy157.
93. Xu M, Guo L, Gu S, Wang O, Zhang R, Peters BA, Fan G, Liu X, Xu X,
Deng L et al. TGS-​GapCloser: A fast and accurate gap closer for large
genomes with low coverage of error-​prone long reads. GigaScience 2020,
9(9):giaa094.
94. Zimin AV, Salzberg SL. The SAMBA tool uses long reads to improve the con-
tiguity of genome assemblies. PLoS Comput Biol 2022, 18(2):e1009860.
95. Schmeing S, Robinson MD. Gapless provides combined scaffolding, gap
filling and assembly correction with long reads. bioRxiv 2022, doi: https://​doi.
org/​10.1101/​2022.03.08.483​466
De Novo Genome Assembly with Long and/​or Short Reads 291

96. Simao FA, Waterhouse RM, Ioannidis P, Kriventseva EV, Zdobnov EM.
BUSCO: assessing genome assembly and annotation completeness with
single-​copy orthologs. Bioinformatics 2015, 31(19):3210–​3212.
97. Gurevich A, Saveliev V, Vyahhi N, Tesler G. QUAST: quality assessment tool
for genome assemblies. Bioinformatics 2013, 29(8):1072–​1075.
98. Alonge M, Soyk S, Ramakrishnan S, Wang X, Goodwin S, Sedlazeck FJ,
Lippman ZB, Schatz MC. RaGOO: fast and accurate reference-​ guided
scaffolding of draft genomes. Genome Biol 2019, 20(1):224.
99. Tamazian G, Dobrynin P, Krasheninnikova K, Komissarov A, Koepfli KP,
O’Brien SJ. Chromosomer: a reference-​based genome arrangement tool for
producing draft chromosome sequences. GigaScience 2016, 5(1):38.
100. Kim J, Larkin DM, Cai Q, Asan, Zhang Y, Ge RL, Auvil L, Capitanu B, Zhang
G, Lewin HA et al. Reference-​assisted chromosome assembly. Proc Natl Acad
Sci U S A 2013, 110(5):1785–​1790.
101. Nurk S, Koren S, Rhie A, Rautiainen M, Bzikadze AV, Mikheenko A, Vollger
MR, Altemose N, Uralsky L, Gershman A et al. The complete sequence of a
human genome. Science 2022, 376(6588):44–​53.
13 
Mapping Protein-​DNA Interactions
with ChIP-​Seq

13.1 Principle of ChIP-​Seq
Without the involvement of DNA-​ interacting proteins, the information
coded in DNA cannot be accessed, transcribed, and maintained. Besides a
large number of transcription factors and coactivators, key DNA-​interacting
proteins include histones, DNA and RNA polymerases, and enzymes for DNA
repair and modification (e.g., methylation). Through their DNA-​interacting
domains, such as helix-​turn-​helix, zinc finger, and leucine zipper domains,
these proteins interact with their DNA targets by hydrogen bonding, hydro-
phobic interactions, or base stacking. Because the intimate relationship
between DNA and these proteins plays an important role in the functioning
of the genome, studying how proteins and DNA interact and where DNA-​
interacting proteins bind across the genome provides key insights into the
many roles these proteins play in various aspects of genomic function,
including information exposure, transcription, and maintenance.
ChIP-​seq is an NGS-​based technology to locate binding sites of a DNA-​
interacting protein in the genome. An exemplary scenario in using ChIP-​
seq is to study transcription factor binding profiles in the genome under
different conditions, such as development stages or pathological conditions.
To achieve this, the protein of interest is first cross-​linked covalently to DNA
in cells with a chemical agent, usually formaldehyde (Figure 13.1). Then the
cells are disrupted, and subsequently sonicated or enzymatically digested to
shear chromatin into fragments that contain 100–​300 bp DNA, followed by
enrichment of the target protein with its bound DNA by immunoprecipitation
using an antibody specific for the protein. Subsequently, the enriched
protein-​DNA complex is dissociated by reversing the cross-​links previously
formed between them, and the released DNA fragments are subjected to
NGS. One key experimental factor in the ChIP-​seq process is the quality of
the antibody used in the enrichment step, as the use of a poor-​quality anti-
body can lead to high experimental noise due to non-​specific precipitation
of DNA fragments.
Based on the size the region that they bind, DNA-​interacting proteins can
be divided into three groups: (1) punctate-​binding: these proteins, usually

DOI: 10.1201/9780429329180-16 293


294 Next-Generation Sequencing Data Analysis

FIGURE 13.1
The basic steps of ChIP-​seq. (From AM Szalkowski, CD Schmid, Rapid innovation in ChIP-​seq
peak-​calling algorithms is outdistancing benchmarking efforts, Briefings in Bioinformatics 2011,
12(6):626–​33. With permission.)
Mapping Protein-DNA Interactions with ChIP-Seq 295

transcription factors, bind to a genomic region that is a few hundred bp or


less in size; (2) broad-​binding: chemically modified histones (such as histone
H3 lysine 27 trimethylation or H3K27me3, and H3 lysine 79 dimethylation
or H3K79me2), or other proteins associated with chromatin domains, bind to
a much larger area of the genome up to several hundred thousand bp; and
(3) mixed or intermediate-​binding: these include some histone modifications
(such as H3 lysine 27 acetylation or H3K27ac, and H3 lysine 4 trimethylation
or H3K4me3), and proteins such as RNA polymerase II, which bind to regions
of the genome that are a few thousand bp in size.

13.2 Experimental Design
13.2.1 Experimental Control
Appropriate control for a ChIP-​seq experiment is key to accounting for
artifacts or biases that might be introduced into the experimental process.
These artifacts and biases may include potential antibody cross-​reactivity
with non-​specific protein factors, higher signal from open chromatin regions
(since they are easier to be fragmented than closed regions [1]), and uneven
sequencing of captured genomic regions due to variations in base compos-
ition. Two major types of controls are usually set up for ChIP-​seq signal adjust-
ment. One is input control, i.e., chromatins extracted from cells or tissues,
which are subjected to the same cross-​linking and fragmentation procedure
but without the immunoprecipitation process. The other is “mock” control,
which is processed by the same procedure including immunoprecipitation;
the immunoprecipitation, however, is carried out using an irrelevant anti-
body (e.g., IgG). While it may seem to serve as the better control between
the two, the “mock” control often produces much less DNA for sequencing
than real experimental ChIP samples. Although sequencing can be carried
out on amplified DNA in this circumstance, the amplification process adds
additional artifact and bias to the sequencing data, which justifies the use of
input DNA as experimental control in many cases.

13.2.2 Library Preparation
To prepare ChIP DNA for library prep, 1–​10 million cells are typically needed
[2]. Within this range, studies of broad-​ binding protein factors require
less cells than those of punctate-​binding proteins. In terms of the amount
of immunoprecipitated DNA required for library prep, while it is often
suggested to start with 5–​10 ng ChIP DNA, it is common to obtain less DNA,
296 Next-Generation Sequencing Data Analysis

which often still generates high-​quality libraries for sequencing. For library
prep, the steps involved include end repair, A-​tailing, incorporation of 3’ and
5’ adapter sequences, size selection, and PCR amplification for final library
generation. The number of cycles used in the PCR amplification step is
important, and overamplification should be avoided as it can affect fragment
representation and library complexity. If obtaining large numbers of cells
is challenging or not feasible, alternative methods such as CUT&RUN (or
Cleavage Under Targets and Release Using Nuclease), which can generate
high-​quality data from as low as 100 cells due to its low background [3], can
be employed.

13.2.3 Sequencing Length and Depth


Single-​end 50–​75 base reads are typically used for ChIP-​seq libraries [4].
Paired-​end sequencing can also be used to help increase mapping, especially
for proteins that may bind to repetitive regions of the genome, but is not
required in most cases. How many reads to obtain for a ChIP-​seq experiment
depends on the size of the genome and how many binding sites the pro-
tein of interest has in the genome. A good indication of having reached suf-
ficient sequence depth is when the number of protein binding sites reaches
plateau with increasing numbers of reads. As a practical guide, for analyzing
a transcription factor that has thousands of binding sites in the mammalian
genome, 20 million reads may be sufficient. Fewer reads may suffice for a
smaller genome, while more reads are required for proteins that bind to the
genome at a higher frequency or with larger “footprint.” To locate binding
regions of these proteins, including histone marks, 30–​50 million reads might
be needed for a genome at the scale of the human genome [5]. Higher sequen-
cing depth is required for control samples in order to obtain background
signals from most regions of the genome. If using CUT&RUN when starting
from low numbers of cells, paired-​end sequencing is suggested, but short
read length (e.g., 25 x 25 bases) at a depth of 5 million read pairs per sample
is usually sufficient [3].

13.2.4 Replication
To examine the reproducibility of a ChIP-​seq experiment, and also reduce
FDR, replicate samples should be used. If a protein of interest binds to
regions of the genome with high affinity, the bound regions should be iden-
tified in replicate samples. Regions that are not identified in replicates are
most possibly due to experimental noise. Pearson correlation coefficient
(PCC) between biological replicates serves as a measurement of experi-
mental reproducibility, while irreproducible discovery rate (IDR) is another
such metric. The calculation and usage of PCC and IDR are detailed later in
this chapter.
Mapping Protein-DNA Interactions with ChIP-Seq 297

13.3 Read Mapping, Normalization, and Peak Calling


13.3.1 Data Quality Control and Read Mapping
The first step in ChIP-​seq data analysis (Figure 13.2) is to evaluate reads
quality. The quality control (QC) metrics detailed in Chapter 5 need to be
examined. If necessary, low-​quality reads should be filtered out and low-​
quality bases trimmed off. Other aspects of determining ChIP-​ seq data
quality include assessing library complexity and experimental reproducibility
between replicates. Assessment of library complexity is important, as low-​
complexity libraries, caused by limited starting material, over-​crosslinking,
low antibody quality, or PCR over-​amplification, can lead to skewed reads
distribution. Library complexity, defined as the proportion of non-​redundant
reads, can be examined with tools such as Preseq [6], or using PCR bottleneck
coefficient (PBC), which is the ratio of N1/​Nd, with N1 being the number

Experimental Design
(Controls, sequencing depth, replication)

Data QC

Read Mapping

Peak Calling

Peak Visualization

Differential Binding Analysis

Functional Analysis
(Peaks assigned to nearby genes)

DNA-Binding Motif Analysis

FIGURE 13.2
Basic ChIP-​seq data analysis workflow.
298 Next-Generation Sequencing Data Analysis

of non-​redundant, uniquely mapped reads, and Nd the number of uniquely


mapped reads. PBC is calculated by a component of ENCODE Software Tools
[7] called phantompeakqualtools, which, besides PBC, also calculates other
quality metrics, i.e., normalized strand cross-​correlation (NSC) and relative
strand cross-​correlation coefficients (RSC), as measures of sequence enrich-
ment (NSC and RSC will be discussed more in the subsequent section on
“Peak Calling”). The assessment of experimental reproducibility is usually
performed by analyzing IDR, which can be calculated using another compo-
nent of ENCODE Software Tools called “Irreproducible Discovery Rate
(IDR)” [8].
The assessment of library complexity and experimental reproducibility
by the ENCODE Software Tools, or the use of other ChIP-​seq QC tools
such as CHANCE [9], deepTools2 [10], and SSP [11], requires mapping
the filtered/​trimmed reads to a reference genome. For this mapping, the
mappers introduced in Chapter 5, including Bowtie2 or BWA, can be used.
One mapping parameter that directly affects subsequent binding site detec-
tion sensitivity and specificity is whether to use multireads, i.e., reads that
map to multiple genomic regions. Multireads may represent background
noise and, if this is the case, should be excluded from further analysis, but
they may also represent true signals located in repeats or duplicated regions.
Including them increases sensitivity but at the expense of higher FDR, while
excluding them improves specificity but at the risk of losing true signals. The
choice for their inclusion or not, therefore, is dependent on whether sensi-
tivity or specificity is given priority. Independent of whether multireads are
used or not, the fraction of uniquely mapped reads among all reads, also
called the non-redundant fraction (NRF), is indicative of data quality. Per
ENCODE guidelines, the NRF should be at least 0.8 [12]. If it drops below
0.5, it indicates concerning problem(s) with the experimental procedure and
caution should be used in the interpretation of the data.
For ChIP-​seq read mapping, it is also worth mentioning that ChIP is an
enrichment, not purification, of protein-​bound DNA sequences. As a result,
more reads are usually generated from background noise than from bound
regions. The background noise can be determined empirically with the use
of input control samples. The distribution of observed background noise is
not random as many would expect (Figure 13.3). Instead, it is affected by
the density of mappable reads in different genomic regions, and the local
chromatin structure (e.g., as mentioned previously, open chromatin struc-
ture generates more background reads). True binding signals in ChIP-​seq
samples are usually superimposed on the background noise. At the absence
of input control samples, although the background noise could be estimated
from modeling of the ChIP-​seq data itself, the estimation cannot fully capture
the inherent complexity of the background noise and therefore experimental
controls should always be run. To further complicate the situation, the degree
of protein-​ binding sequence enrichment may also vary from location to
newgenrtpdf
100

* * *

Mapping Protein-DNA Interactions with ChIP-Seq


80
60
40
Pol II ChIP-seq
20
0
100
Overlapping mapped DNA fragments

80
60
40
20
Input DNA
0
100
80
60
40 Interferon- γ–stimulated STAT1 ChIP-seq
20
0
100
80
60
40 Interferon- γ–stimulated input DNA
20
0
1
0.8
0.6
0.4
0.2 Mappable bases (1 Kb)
0

XPNPEP3 R BX1 EP 300

SLC25A17 ST13
FIGURE 13.3
Background noise and signal profiles in a ChIP-​seq experiment. Shown here is the density of mapped reads in one region of the human chromosome 22
for RNA polymerase II and the transcription factor STAT1 (tracks 1 and 3, count from the top), respectively. Genes coded by the two DNA strands in this
region are displayed at the bottom. Tracks 2 and 4 show the distribution of mapped reads for the respective input DNA controls for the two proteins. It
should be noted that some of the peaks in the protein tracks are also present in their input controls. Track 5 displays the fraction of uniquely mappable

299
bases. (Adapted by permission from Macmillan Publishers Ltd: Nature Biotechnology, PeakSeq enables systematic scoring of ChIP-​seq experiments
relative to controls, J Rozowsky, G Euskirchen, RK Auerbach, ZD Zhang, T Gibson, R Bjornson, N Carriero, et al., copyright 2009.)
300 Next-Generation Sequencing Data Analysis

location, with some having strong signals while others more modest signals.
The degree of enrichment at each location is not necessarily a reflection of
their biological importance, as those with more modest enrichment may be
equally important as those at the top of the enrichment list.
After mapping, reproducibility between replicate samples and overall simi-
larity between different samples can be examined using established tools. For
example, the multiBamSummary tool in deepTools2 can be used to check
reads coverage across the entire genome in “bins” mode from two or more
input BAM files, and another tool called plotCorrelation can take the output
to compute and visualize sample correlations. The plotFingerprint tool in
the same toolset can be used to visualize cumulative reads enrichment as an
indication of target DNA enrichment efficiency. This tool is most informative
for punctate-​binding proteins such as transcription factors. Besides sample
correlations and the other aforementioned QC measures such as PBC, add-
itional QC analyses can also be performed. For example, visualization of the
distribution of mapped reads in the genome, using the visualization tools
introduced in Chapter 5, can offer further clues on data quality. This is espe-
cially true when some specific binding regions have already been known for
the protein of interest. In comparison to those from input control samples,
sequence reads from ChIP samples should show strong clustering in these
regions.

13.3.2 Peak Calling
Peak calling, the process of finding regions of the genome to which the pro-
tein of interest binds, is a key step in ChIP-​seq data analysis. It is basically
achieved through locating regions where reads are mapped at levels signifi-
cantly above the background. As this process is also applicable to ATAC-​seq
(assay for transposase-​accessible chromatin using sequencing) and DNase-​
seq (DNase I hypersensitive sites sequencing), both of which aim to locate
transcriptionally accessible regions of the genome, many of the methods
introduced below can also be used for analysis of ATAC-​seq and DNase-​
seq data. Among currently available peak calling methods, the simplest is
to count the total number of reads mapped along the genome, and call each
location with the number of mapped reads over a threshold as a peak. Due to
the inherent complexity in signal generation, including uneven background
noise and other confounding experimental factors, this approach is overly
simplistic. Among the experimental factors, the way the immunoprecipitated
DNA fragments are sequenced on most platforms has a direct influence on
how peaks are called. Since the reads are usually short, only one end or both
ends of a fragment, depending on whether single-​end or paired-​end reads
are produced, are sequenced. To locate a target protein’s binding regions,
which are represented by the immunoprecipitated DNA fragments and
not just the generated reads, peak calling algorithms need to either shift or
extend the reads to cover the actual binding areas. For example, MACS2
Mapping Protein-DNA Interactions with ChIP-Seq 301

shifts reads mapped to the two opposite strands toward the 3’ ends, based on
the average DNA fragment length, to cover the most likely protein binding
location [13]. SPP uses a similar strategy to shift each read mapped to either
strand relative to each other until strand cross-​correlation coefficient reaches
the highest level at the shift that equals to the average length of the DNA
fragments (Figure 13.4) [14]. PeakSeq uses an alternative approach to extend
the reads in the 3’ direction to reach the average length of DNA fragments in
the library [15].
The reads shift approach and the strand cross-​correlation profile shown in
Figure 13.4 can also be used to evaluate ChIP-​seq data quality. When using
short reads (usually less than 100 bases) to analyze large target genomes,
which usually results in a significant number of reads being mapped to mul-
tiple genomic locations, a “phantom” peak also exists at a shift that equals to
the read length (Figure 13.5). If a run is successful, the fragment-​length ChIP
peak should be significantly higher than the read-​length “phantom” peak,
as well as the background signal. The aforementioned ENCODE software
phantompeakqualtools provides two indices for the examination of strand
cross-​correlation, i.e., NSC, being the ratio of the cross-​correlation coefficient
at the fragment-​length peak over that of the background, and RSC, as the
ratio of background-​adjusted cross-​correlation coefficient at the fragment-​
length peak over that at the “phantom” peak.
Shifting reads mapped to the positive and negative strands toward the
center, or extending reads to reach the average fragment length, in order to
count the number of aggregated reads at each base pair position is the first
sub-​step to peak calling. As illustrated in Figure 13.6, peak calling involves
multiple substeps. First, a signal profile is created through smoothing of
aggregated read count across each chromosome. Subsequently, background
noise needs to be defined and the signals along the genome need be adjusted
for the background. One simple approach is to subtract read counts in con-
trol sample, if available, from the signal across the genome, or use the signal/​
noise ratio. Subsequently, to call peaks from the background-​adjusted ChIP-​
seq signal, often-​used approaches include using absolute signal strength,
signal enrichment in relation to background noise, or a combination of
both. To facilitate determination of signal enrichment, statistical signifi-
cance is often computed using Poisson or negative binomial distributions.
Empirical estimation of false discovery rate (FDR) can be carried out by first
calling peaks using control data (i.e., false positives), and subsequently cal-
culating the ratio of peaks called from the control to those called from the
ChIP sample. Alternatively, the Benjamini–Hochberg approach introduced in
Chapter 7 can also be applied to correct for multiple comparisons. After peak
calling, artifactual peaks need to be filtered out, including those that contain
only one or a few reads that are most possibly due to PCR artifacts, or those
that involve significantly imbalanced numbers of reads on the two strands
(see Figure 13.6).
newgenrtpdf
302
60 0.14
Protein 10
40 0.12

Strand cross-correlation coefficient


+
5 0.10
– 20

Tag counts
Tag density
0.08
0 0
0.06

–20 0.04
–5
Crosslink 0.02
Fragmentation –40
Positive-strand tag –10
0.00
Negative-strand tag –100 –50 0 50 100 150 200 250 300
110,500,100 110,500,200 110,500,300
Position on chromosome 1 Relative strand shift

Next-Generation Sequencing Data Analysis


(a) (b) (c)
FIGURE 13.4
The distribution of ChIP-​seq reads around the actual binding region and their positional shift on the two DNA strands. (a) How ChIP-​seq reads
are produced from cross-​linked and fragmented DNA. The cross-​linking between the protein and the bound DNA can occur at different sites,
as is the fragmentation of the DNA. Each fragment is read at its 5’ end (indicated by the squares). These reads, serving as sequence tags of each
fragment, are clustered around the actual binding region from the two sides depending on which strand they come from. The dashed red line
depicts a fragment from a long cross-​link. (b) The distribution of sequence tag signal around the binding region. Vertical lines represent counts
of sequence tags whose 5’ end maps to each nucleotide position on the positive (red) and negative (blue) strands. The solid curves represent
smoothed tag density along each strand. Since the two curves approach the binding site from the two sides, there is a gap between their peaks.
(c) Strand cross-​correlation associated with shifting the strands across the gap. Before shifting the strands, Pearson correlation coefficient is
calculated between tag density of the two strands. When sequence tags mapped to the two strands are shifted relative to each other (shown
on the x-​axis), the Pearson correlation coefficient gradually changes (y-​axis). The dashed gray line at x=​0 corresponds to the strand cross-​
correlation before the shift, while the dashed red line at the peak to the highest cross-​correlation coefficient at the strand shift that equals to the
average length of the DNA fragments. (Adapted by permission from Macmillan Publishers Ltd: Nature Biotechnology, Design and analysis of
ChIP-​seq experiments for DNA-​binding proteins, PV Kharchenko, MY Tolstorukov, PJ Park, copyright 2008.)
newgenrtpdf
Mapping Protein-DNA Interactions with ChIP-Seq 303
newgenrtpdf
FIGURE 13.5

304
The “phantom” peak and its use in determining ChIP-​seq data quality. The “phantom” peak corresponds to the cross-​
correlation at the strand shift that equals to the read length, while the ChIP peak corresponds to the cross-​correlation
at the shift of the average DNA fragment length. A successful run is characterized by the existence of a predominant
ChIP peak and a much weaker “phantom” peak. In marginally passed or failed runs, the former diminishes while the
latter relatively becomes much stronger. (Adapted from SG Landt, GK Marinov, A Kundaje, P Kheradpour, F Pauli, S
Batzoglou, BE Bernstein et al., ChIP-​seq guidelines and practices of the ENCODE and modENCODE consortia, Genome
Research 2012, 22(9):1813–​1831. Used under the terms of the Creative Commons License (Attribution-​NonCommercial
3.0 Unported License) as described at http://​crea​tive​comm​ons.org/​licen​ses/​by-​nc/​3.0/​. ©2012 Cold Spring Harbor
Laboratory Press.)

Next-Generation Sequencing Data Analysis


Mapping Protein-DNA Interactions with ChIP-Seq 305

FIGURE 13.6
Basic substeps of calling peaks from ChIP-​seq data. The P(s) at bottom left signifies the probability
of observing a location covered by S mapped reads, and the sthresh marks the threshold for calling
a peak significant. Bottom right shows two types of possible artifactual peaks: single strand
peaks and those based on mostly duplicate reads. (Adapted by permission from Macmillan
Publishers Ltd: Nature Methods, Computation for ChIP-​seq and RNA-​seq studies, S Pepke, B
Wold, A Mortazavi, copyright 2009.)
306 Next-Generation Sequencing Data Analysis

For implementation of this peak calling process, different peak callers use
different methods, which can lead to differences in final outcomes. Among
currently available peak callers, MACS2, HOMER (findPeaks module) [16],
SICER2 [17], JAMM [18], SPP, and PeakSeq are among some of the popular
ones (see Table 13.1). Among these methods, MACS2 uses Poisson distribu-
tion to model reads distribution across the genome. To achieve robust peak
prediction, this modeling process considers the dynamic nature and effects
of local background noise as caused by biological factors such as local chro-
matin structure and genome copy number variations, and technical biases
introduced during library prep, sequencing, and mapping processes. Peaks
are called from enrichment of reads in a genomic region with statistical sig-
nificance calculated based on Poisson distribution. On FDR estimation, while
the original MACS uses an empirical approach through exchanging ChIP
and control samples, MACS2 applies the Benjamini–Hochberg method to
adjust p-​values. The findPeaks module in HOMER also uses Poisson distri-
bution to identify putative peaks. To arrive at a list of high-​quality peaks,
different filters are then applied to these putative peaks. These filters are
either based on the use of (1) control samples, i.e., peaks need to pass fold-​
change and cumulative Poisson p-​value thresholds in comparison to control
samples; (2) local read counts, i.e., the density of reads at a peak need to be

TABLE 13.1
A Short List of ChIP-​Seq Peak Calling Algorithms
Name Description Reference

MACS/​MACS2 Empirically models ChIP-​seq read length to improve peak [13]


prediction, uses a dynamic Poisson distribution
HOMER Identifies peaks based on the principle that more [16]
(findPeaks sequencing reads are found in these regions than
module) expected by chance
PeakSeq Based on a two-​pass strategy to compensate for open [15]
chromatin signal
SPP Includes binding profile normalization, peak detection, [14]
and estimation of read depth to achieve peak saturation
CisGenome Features multifaceted interactive analysis and customized [21]
batch-​mode computation
SICER2 Uses a clustering approach to identify enriched domains [17]
from histone modification ChIP-​seq data
SiSSRs Uses the direction and density of reads and the average [22]
DNA fragment length to identify binding sites
PeakRanger Applies a staged algorithm to discover enriched regions [23]
and the summits within them
ZINBA Models and accounts for factors co-​varying with [24]
background or true signals
Mapping Protein-DNA Interactions with ChIP-Seq 307

significantly higher than that in the surrounding region; or (3) clonal signal,
to remove peaks with high number of reads that map to only a small number
of unique positions. SPP, an R package, calculates smoothed read enrichment
profile along the genome and identifies significantly enriched sites compared
to input control using methods such as WTD (Window Tag Density). The
WTD method considers sequence tag patterns immediately upstream and
downstream of the center of the binding location, thereby increasing predic-
tion accuracy. The peak calling employed in PeakSeq is a two-​pass process. In
its first pass, PeakSeq uses background modeling to identify initial potential
binding regions. To adjust for background using control data, the fraction of
reads located in the initially identified potential binding regions are excluded
and the reads in the remainder of the genome in the ChIP-​seq sample are
normalized to the control data by linear regression. In the second pass, target
peaks are called by scoring reads-​enriched target regions based on calcula-
tion of fold enrichment in ChIP-​seq sample vs. control, and statistical signifi-
cance associated with each enriched target region is calculated from binomial
distribution. More recently, newer algorithms based on the application of
deep learning approaches have begun to emerge, such as CNN-​Peaks [19]
and LanceOtron [20].
To ensure the robustness of analysis results, it is recommended to use more
than one method for peak calling. While IDR is usually used to measure the
rate of irreproducible discoveries, i.e., peaks that are called in one replicate
sample but not in another, it can also be used to compare peak calling results
generated from different methods. The original use of IDR in assessing rep-
licate reproducibility is based on the rationale that peaks of high significance
are more consistently ranked across replicates and therefore have better
reproducibility than those of low significance. As shown in Figure 13.7, to
compare a pair of ranked lists of peaks identified in two replicates, IDR are
plotted against the total numbers of ranked peaks. Since IDR computation
relies on both high significance (more reproducible) and low significance
(less reproducible) peaks, peak calling stringency needs to be relaxed to
allow generation of both high and low confidence calls. The transition in
this plot from reliable signal to noise is an index of overall experimental
reproducibility. Because IDR is independent of any particular peak-​calling
method, it can be applied to compare the performance of different peak
calling methods on a particular dataset and therefore help pick the most
appropriate method(s) (Figure 13.8). IDR can also be used to evaluate repro-
ducibility across experiments and labs.

13.3.3 Post-​Peak Calling Quality Control


After peaks are called, prior to conducting further downstream analyses, it
is a good practice to perform another QC step to determine the quality of
peaks called. Tools such as deepTools2 and ChIPQC [25] can be used for this
308 Next-Generation Sequencing Data Analysis

FIGURE 13.7
Use of irreproducible discovery rate (IDR) in assessing replicate reproducibility. Panel (A) shows
the distribution of the significance scores of the peaks identified two replicate experiments. The
IDR method computes the probability of being irreproducible for each peak, and classifies them
as being reproducible (black) or irreproducible (red). Panel (B) displays the IDR at different
rank thresholds when the peaks are sorted by the original significance score. (From T Bailey, P
Krajewski, I Ladunga, C Lefebvre, Q Li, T Liu, P Madrigal, C Taslim, J Zhang, Practical guidelines
for the comprehensive analysis of ChIP-​seq data, PLoS Computational Biology 2013, 9:e1003326.
Used under the terms of the Creative Commons Attribution License (http://​crea​tive​comm​ons.
org/​licen​ses/​by/​3.0/​). © 2013 Bailey et al.)

0.5 MACS
CisGenome
0.4 spp
SISSRs
0.3 Useq
IDR

QuEST
0.2
0.1
0
0

0
50

00

50

00

50
1,

1,

2,

2,

Number of peaks
FIGURE 13.8
Evaluation of the performance of six peak callers using IDR. (Adapted by permission from
Macmillan Publishers Ltd: Nature Methods, Systematic evaluation of factors influencing ChIP-​seq
fidelity, Y Chen, N Negre, Q Li, JO Mieczkowska, M Slattery, T Liu, Y Zhang et al., copyright 2012.)

step. For example, to check for consistency of called peaks between replicate
samples, the aforementioned multiBamSummary tool in deepTools2 can
be used in “BED-​file” mode using BED files from peak callers as input, and
the output passed on to plotCorrelation to generate visualization of samples
based on their correlation coefficients.
ChIPQC reports a number of quality metrics related to called peaks,
including peak signal strength, enrichment of reads in peaks, and relative
Mapping Protein-DNA Interactions with ChIP-Seq 309

enrichment of peaks in various genomic features. The metrics related to


peak signal strength are relative strand cross-​correlation coefficient, i.e.,
RSC, and estimated fragment length, which should be very close to the
DNA fragment length produced during library prep. Enrichment of reads in
peaks is represented by metrics including the percentage of reads in peaks,
standardized standard deviation (SSD) of genomic coverage, and the per-
centage of reads in ENCODE blacklisted regions. Among these metrics, the
percentage of reads in peaks, also often termed FRiP (or fraction of reads
in peaks), is an overall indicator of immunoenrichment and ChIP-​seq data
quality. Usually only a small percentage of reads map to peak regions, and
the majority of reads only represent background. As a general guideline, the
ENCODE Consortium sets 1% as the minimum for an acceptable FRiP with
MACS as the peak caller using default parameters. As they can vary with the
use of different peak callers and parameters, FRiP values must be derived
from the same algorithm using same parameters in order for them to be
comparable across samples or experiments. The SSD metric is a measure
of variability of reads genomic coverage. If reads are highly enriched in
peak regions, as expected from a well-​executed ChIP-​seq experiment, the
SSD values associated with ChIP samples should be much higher than
those with control samples. The percentage of reads in blacklisted regions is
another metric that affects peak calling outcome. Since some regions of the
genome, especially highly repetitive regions, can produce artifactually high
enrichment signals independent of experiment, the ENCODE Consortium
has compiled a blacklist for such regions [26]. Since reads mapped to the
blacklisted regions often confound peak calling and fragment length estima-
tion, they should be monitored and filtered out. Lastly, on relative enrich-
ment of peaks in various genomic features, the distribution of reads across
major features including promoters, 5’ UTRs, coding regions, introns, and
3’ UTRs, provides another evaluation of ChIP-​seq signal distribution. For
instance, transcription factors are expected to have the highest enrichment
levels in promoters and 5’ UTRs.

13.3.4 Peak Visualization
Visualizing peaks in their genomic context allows identification of overlap-
ping or nearby functional elements, and thereby facilitates peak annotation
and data interpretation. Many peak callers generate BED files containing
peak chromosomal locations, along with WIG and bedGraph track files,
all of which can be uploaded to a genome browser for peak visualization.
Examination of peak regions in a genome browser and comparison with
other data/​annotation tracks allow identification of associated genomic
features, such as promoters, enhancers, and other regulatory regions.
BEDTools can also be applied to explore relationships between peaks
and other genomic landmarks such as nearby protein-​ coding or non-​
coding genes.
310 Next-Generation Sequencing Data Analysis

13.4 Differential Binding Analysis


Binding of DNA-​interacting proteins to their target genomic regions is a
quantitative process, that is, they occupy these regions at different rates
under different conditions. This is due to regional accessibility, presence/​
absence of other protein partners, and/​or other factors that regulate their
binding. Differential binding analysis answers the question of how a target
protein changes its DNA-​binding pattern under different conditions. There
are two different approaches for this analysis, with one qualitative and
the other quantitative. The qualitative approach compares peaks called in
different conditions, and divides them into “shared” and “unique” [27].
This approach is simple, but it does not use the quantitative information
generated in the peak calling process, so it is best used to produce a rough
initial estimation of differential binding. The quantitative approach, through
analysis of read counts or read densities in peak regions or on a sliding
window basis across the genome, generates statistical assessment of the
degree of differential binding between conditions. As this is similar to the
RNA-​seq-​based differential expression analysis despite the higher level of
background noise, data normalization is required to adjust for systematic
biases that are unrelated to biological factors. For the comparison of two or
more ChIP-​seq samples, such biases include immunoprecipitation efficiency
and sequencing depth.
Similar to normalizing RNA-​seq data, adjusting for sequencing depth is the
simplest approach. In this approach, the total numbers of reads in different
samples are adjusted, by multiplying a scaling factor to each sample, to a
same target level, e.g., the median or lowest total read count among the
samples. The basic assumption for this approach is that the overall number
of binding sites for a target protein does not change across different experi-
mental conditions. Although this approach is simple and straightforward, it
does not take into consideration the difference in signal-​to-​noise ratio that
is often observed in different samples. If one sample library is noisier and
contains more background reads, these reads, while not representing true
signals, are still counted into the total read number. This situation will there-
fore lead to bias in the normalized data.
There are several currently available normalization approaches that con-
sider this issue of signal-​to-​noise ratio variation among samples. For example,
the normalization procedure used in diffReps first identifies and removes
regions with low read count (mostly background noise) [28]. The subsequent
normalization is then based on the remaining regions, using a linear pro-
cedure similar to that used by DESeq2. Another similar approach uses only
reads mapped to peaks. In this modified sequencing depth-​based normal-
ization approach, the total number of reads mapped to the peak regions are
used as the basis for calculating scaling factor for each sample [29]. Using
Mapping Protein-DNA Interactions with ChIP-Seq 311

this approach, the normalized peak signal is computed as the original peak
sequence read count being scaled by the sum of read counts of all peaks, i.e.,

Xi , j
Zi , j =

N
j =1
Xi , j

where Zi , j and Xi,j are the normalized and original peak signal for sample i
and peak j, and N is the total number of called peaks.
Normalization methods that were previously developed for microarray
data have also been adapted for ChIP-​seq data. MAnorm2, as an example,
uses a hierarchical normalization process that is similar to the MA plot
approach used for microarray data. Based on the assumption that signals
from common peaks shared among all samples remain unchanged globally,
MAnorm2 applies a linear transformation process to the raw read counts
in order to make both the arithmetic mean of M values (or differences in
signals between samples), and the covariance between M and A (average
signals between two samples) values, to be zero [30]. ChIPnorm uses a
modified version of quantile normalization [31]. A locally weighted regres-
sion (LOESS) normalization approach for ChIP-​seq data [32] is similar to the
LOESS procedure applied to cDNA microarray data normalization. All these
approaches assume that the overall binding profile of the target protein does
not vary across different conditions. This assumption does not hold, however,
under conditions when the overall level or activity of the protein under study
changes due to experimental perturbation. Under such conditions, normal-
ization approaches based on spike-​in of an exogenous reference epigenome
at a constant amount [33], similar to the use of spike-​in RNA controls in bulk
RNA-​seq, can be used.
Besides all the normalization approaches introduced above, good experi-
mental design and consistent experimental procedure can minimize data
variability in different samples and groups, thereby alleviating the burden
on posterior normalization. For example, processing all samples side by side
using the same experimental procedure and parameters, such as the same
antibody, by the sample operator, will minimize sample-​ to-​
sample vari-
ability. When conducting an experiment in this way, the simpler normaliza-
tion approach based on total library read count can be sufficient.
Since the ChIP-​seq-​based quantitative analysis of differential binding is
similar to the RNA-​ seq-​
based differential expression analysis, packages
such as edgeR and DESeq2 can be applied here. Table 13.2 lists some of the
packages that are designed for ChIP-​seq differential binding analysis. As
listed these packages can be divided into two categories, with one composed
of methods that are dependent on peak calling from an external application,
while the other of those that handle peak calling internally or do away with
peak calling altogether. For methods that require internally or externally
312 Next-Generation Sequencing Data Analysis

TABLE 13.2
Packages Developed for ChIP-​Seq Differential Binding Analysis
Name Description Reference

Methods that require externally called peaks


DiffBind Uses statistical tests used in RNA-​seq packages edgeR and [38]
DESeq to process peak sets and identify differentially
bound regions
ChIPComp Differential binding analysis taking into considerations of [42]
controls, signal-​to-​noise ratios, replicates, and multi-​factor
experimental design
MAnorm2 Conducts a hierarchical MA-​plot-​based normalization prior [30]
to differential analysis
DBChIP Identifies differentially bound punctate binding sites in [39]
multiple conditions using RNA-​seq DE approaches and
accommodates controls
DIME Differential binding analysis using a finite exponential-​ [43]
normal mixture model
MMDiff2 Takes a multivariate non-​parametric approach to test [44]
differential binding
Methods that do not rely on pre-​called peaks
bdgdiff/​ A module of MACS2 that detects differential binding based [13]
MACS2 on paired four bedGraph files
diffReps Detects and annotates differential chromatin modification [28]
hotspots using a sliding window approach
ChIPDiff Differential histone mark analysis based on HMM [36]
RSEG Uses HMM to locate differentially bound broad genomic [45]
domains associated with diffusive histone modification
marks
csaw Uses the sliding window approach for detection of [34]
differential binding regions, with different window sizes set
for transcription factors and histone marks
PePr Another method that uses sliding windows, and a negative [35]
binomial distribution model to detect differential peaks
THOR Differential peak calling based on an HMM with a three-​state [37]
topology
ChIPnorm Carries out quantile normalization for differential binding [31]
sites identification

called peaks, robust peak calling is essential to produce quality results. For
those that do not rely on peak calling, differential binding analysis aims to
find significant ChIP-​seq signal changes between conditions throughout the
entire genome. These latter methods can be further divided into two subcat-
egories. One subcategory, exemplified by diffReps [28], csaw [34], and PePr
[35], uses sliding windows, the size of which is typically selected based on the
Mapping Protein-DNA Interactions with ChIP-Seq 313

footprint of the target protein. The other subcategory uses complex segmenta-
tion techniques, such as hidden Markov model (HMM), to first segment
genome into bins and then infer the hidden state of each bin in order to detect
differential protein-​binding sites. ChIPDiff [36] and THOR [37] are examples
of this approach.
To test for differential binding, statistical models based on Poisson or nega-
tive binomial distribution are often used, again similar to RNA-​seq DE ana-
lysis. In fact, methods such as DiffBind [38] and DBChip [39] directly inherit
statistical models from edgeR and DESeq2. In terms of detection targets,
while some methods are specifically designed for punctate-​binding protein
factors (such as DBChip) and some others for more broad marks (such as
SICER2 and RSEG), most of the methods can be used for binding regions
of different sizes. In addition, in terms of handling replicate samples, some
can work with experiments that do not use replicate samples, while others
require replicates. As mentioned earlier, use of replication is suggested,
which usually leads to increased detection precision with a much reduced
number of differential binding regions, but in the meantime at the expense of
reduced sensitivity. To help select appropriate methods for a particular appli-
cation, existing benchmarking studies provide decision trees based on their
comparative testing results [40, 41]. It should also be noted that like those
devised for RNA-​seq-​based differential expression analysis, these packages
are designed based on certain assumptions and therefore the user needs to be
aware of these assumptions and ensure they are fulfilled prior to using them.
For example, MAnorm2 is based on the assumption that there is no global
change in binding at peak regions between conditions.

13.5 Functional Analysis
Often the data gathered from a ChIP-​seq study is used to understand gene
expression regulation and associated biological functions. To conduct func-
tional analysis, peaks are first assigned to nearby genes using tools such
as ChIPseeker [46], GREAT (Genomic Regions Enrichment of Annotations
Tool) [47], and ChIPpeakAnno [48]. While it is debatable on what genes a
peak should be assigned to, a straightforward approach is to assign it to the
closest gene transcription start site. Once peaks are assigned to target genes,
an integrated analysis of ChIP-​seq and gene expression data (more on this
in Section 13.7) can be carried out. Furthermore, Gene Ontology, biological
pathway, gene network, or gene set enrichment analyses can be conducted
using similar approaches as described in Chapter 7. Prior to carrying out
these gene functional analyses, one should also bear in mind that the peak-​to-​
gene assignment process is biased by gene size, as the presence of peak(s) has
a positive correlation with the length of a gene. In addition, the distribution
314 Next-Generation Sequencing Data Analysis

of gene size in different functional annotations such as GO categories is not


uniform, with some categories having excess number of long genes while
others having more short genes. To solve the problems caused by different
gene sizes, methods that adjust for the effects of gene size should be used,
such as ChIP-​Enrich [49].

13.6 Motif Analysis
One of the goals of ChIP-​seq data analysis is to identify DNA-​binding motifs
for the protein of interest. A DNA-​binding motif is usually represented by a
consensus sequence, or more accurately, a position specific frequency matrix.
Figure 13.9 A shows an example of such a DNA-​binding motif, the one bound
by a previously introduced transcription factor NRF2 (see Chapter 2). To iden-
tify motifs from ChIP-​seq data, all peak sequences need to be assembled and
fed into multiple motif discovery tools. Some of the commonly used motif
discovery tools are Cistrome [50], Gibbs motif sampler (part of CisGenome),
HOMER (findMotifs module), MEME-​ChIP [51] as part of the MEME suite
[52], rGADEM [53], and RSAT peak-​motifs [54]. The motif discovery phase
usually ends up with one or more motifs, with one being the binding site of

FIGURE 13.9
The consensus DNA-binding motif of the transcription factor NRF2. Panel (A) shows the currently
known NRF2-​binding motif, while panel (B) displays the result of a de novo motif analysis using
NRF2 ChIP-​seq data. (From BN Chorley, MR Campbell, X Wang, M Karaca, D Sambandan, F
Bangura, P Xue, J Pi, SR Kleeberger, DA Bell, Identification of novel NRF2-​regulated genes by
ChIP-​seq: influence on retinoid X receptor alpha, Nucleic Acids Research 2012, 40(15):7416–​7429,
With permission.)
Mapping Protein-DNA Interactions with ChIP-Seq 315

the target protein and others being that of its partners. The discovered motif(s)
can be compared with currently known motifs catalogued in databases such
as JASPAR [55] to detect similarities, find relationships with other motifs,
or locate other proteins that might bind at or near the peak region as part
of a protein complex. Tools for motif comparison include STAMP [56] and
Tomtom [57]. Motif enrichment analysis can also be carried out to find out
if known motifs are enriched in the peak regions using tools such as AME
[58], CentriMo [59], and SEA [60]. Finally motif scanning and mapping by
tools like FIMO [61] allows visualization of the discovered motif(s) in the
ChIP-​seq peak areas. Some of these tools have been integrated into motif ana-
lysis pipelines, such as the MEME Suite [62], which includes MEME-​ChIP,
Tomtom, CentriMo, SEA, AME, and FIMO.

13.7 Integrated ChIP-​Seq Data Analysis


As genomic functions are to a large degree controlled by concerted binding
of a wide array of DNA-​interacting proteins, integrated analysis of ChIP-​
seq data sets generated for a multitude of these proteins affords new
opportunities to gain a comprehensive overview of the functional states
of a genome and the host cell. As a good example, such an integrated ana-
lysis has led to the discovery of a large number of chromatin states, each
of which display distinct sequence motifs and functional characteristics
[63]. The discovery of these chromatin states was achieved with the use
of a multivariate hidden Markov model on a large collection of ChIP-​seq
data, generated for 38 different histone methylation and acetylation marks,
H2AZ (a variant of histone H2A), RNA polymerase II, and CTCF (a tran-
scriptional repressor).
Besides meta-​analysis of multiple ChIP-​seq data sets, integrated analysis
of ChIP-​seq with other genomics data, such as RNA-​seq data, offers fur-
ther information on genome function and regulation. The majority of pro-
tein factors used in various ChIP-​seq studies are transcription factors and
histones that carry a large array of modified marks, all of which are key
regulators of genome transcription. Coupled analysis of matched ChIP-​seq
and RNA-​seq data augments the utility of both data types, and provides new
insights that cannot be obtained from analyzing either data type alone. To
carry out integrative analysis of ChIP-​seq and RNA-​seq data, Bayesian mixed
and hierarchical models [64, 65] have been used. In addition, tools such as
BETA [66], CEAS [67], and ChIPpeakAnno can also be used to help inves-
tigate the correlation between the DNA-binding profile and regulation of
nearby gene transcription.
316 Next-Generation Sequencing Data Analysis

References
1. Chen Y, Negre N, Li Q, Mieczkowska JO, Slattery M, Liu T, Zhang Y, Kim TK,
He HH, Zieba J et al. Systematic evaluation of factors influencing ChIP-​seq
fidelity. Nat Methods 2012, 9(6):609–​614.
2. Visa N, Jordan-​Pla A. ChIP and ChIP-​Related Techniques: Expanding the
fields of application and improving ChIP performance. Methods Mol Biol 2018,
1689:1–​7.
3. Skene PJ, Henikoff JG, Henikoff S. Targeted in situ genome-​wide profiling
with high efficiency for low cell numbers. Nat Protoc 2018, 13(5):1006–​1019.
4. Meyer CA, Liu XS. Identifying and mitigating bias in next-​generation sequen-
cing methods for chromatin biology. Nat Rev Genet 2014, 15(11):709–​721.
5. Jordán-​Pla A, Visa N. Considerations on experimental design and data ana-
lysis of chromatin immunoprecipitation experiments. Methods Mol Biol 2018,
1689:9–​28.
6. Daley T, Smith AD. Predicting the molecular complexity of sequencing
libraries. Nat Methods 2013, 10(4):325–​327.
7. ENCODE Software Tools (www.encode​proj​ect.org/​softw​are/​)
8. Irreproducible Discovery Rate (IDR) (www.encode​proj​ect.org/​softw​
are/​idr/​)
9. Diaz A, Nellore A, Song JS. CHANCE: comprehensive software for quality
control and validation of ChIP-​seq data. Genome Biol 2012, 13(10):R98.
10. Ramirez F, Ryan DP, Gruning B, Bhardwaj V, Kilpert F, Richter AS, Heyne
S, Dundar F, Manke T. deepTools2: a next generation web server for deep-​
sequencing data analysis. Nucleic Acids Res 2016, 44(W1):W160–​165.
11. Nakato R, Shirahige K. Sensitive and robust assessment of ChIP-​seq read dis-
tribution using a strand-​shift profile. Bioinformatics 2018, 34(14):2356–​2363.
12. Landt SG, Marinov GK, Kundaje A, Kheradpour P, Pauli F, Batzoglou S,
Bernstein BE, Bickel P, Brown JB, Cayting P et al. ChIP-​seq guidelines and
practices of the ENCODE and modENCODE consortia. Genome Res 2012,
22(9):1813–​1831.
13. Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum
C, Myers RM, Brown M, Li W et al. Model-​based analysis of ChIP-​Seq (MACS).
Genome Biol 2008, 9(9):R137.
14. Kharchenko PV, Tolstorukov MY, Park PJ. Design and analysis of
ChIP-​ seq experiments for DNA-​ binding proteins. Nat Biotechnol 2008,
26(12):1351–​1359.
15. Rozowsky J, Euskirchen G, Auerbach RK, Zhang ZD, Gibson T, Bjornson R,
Carriero N, Snyder M, Gerstein MB. PeakSeq enables systematic scoring of
ChIP-​seq experiments relative to controls. Nat Biotechnol 2009, 27(1):66–​75.
16. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, Cheng JX, Murre
C, Singh H, Glass CK. Simple combinations of lineage-​determining transcrip-
tion factors prime cis-​regulatory elements required for macrophage and B cell
identities. Mol Cell 2010, 38(4):576–​589.
17. Zang C, Schones DE, Zeng C, Cui K, Zhao K, Peng W. A clustering approach
for identification of enriched domains from histone modification ChIP-​Seq
data. Bioinformatics 2009, 25(15):1952–​1958.
Mapping Protein-DNA Interactions with ChIP-Seq 317

18. Ibrahim MM, Lacadie SA, Ohler U. JAMM: a peak finder for joint analysis of
NGS replicates. Bioinformatics 2015, 31(1):48–​55.
19. Oh D, Strattan JS, Hur JK, Bento J, Urban AE, Song G, Cherry JM. CNN-​
Peaks: ChIP-​Seq peak detection pipeline using convolutional neural networks
that imitate human visual inspection. Sci Rep 2020, 10(1):7933.
20. Hentges LD, Sergeant MJ, Cole CB, Downes DJ, Hughes JR, Taylor S.
LanceOtron: a deep learning peak caller for genome sequencing experiments.
Bioinformatics 2022, 38(18):4255–​4263.
21. Ji H, Jiang H, Ma W, Johnson DS, Myers RM, Wong WH. An integrated soft-
ware system for analyzing ChIP-​chip and ChIP-​seq data. Nat Biotechnol 2008,
26(11):1293–​1300.
22. Jothi R, Cuddapah S, Barski A, Cui K, Zhao K. Genome-​wide identification of
in vivo protein-​DNA binding sites from ChIP-​Seq data. Nucleic Acids Res 2008,
36(16):5221–​5231.
23. Feng X, Grossman R, Stein L. PeakRanger: a cloud-​enabled peak caller for
ChIP-​seq data. BMC Bioinformatics 2011, 12:139.
24. Rashid NU, Giresi PG, Ibrahim JG, Sun W, Lieb JD. ZINBA integrates local
covariates with DNA-​seq data to identify broad and narrow regions of enrich-
ment, even within amplified genomic regions. Genome Biol 2011, 12(7):R67.
25. Carroll TS, Liang Z, Salama R, Stark R, de Santiago I. Impact of artifact removal
on ChIP quality metrics in ChIP-​seq and ChIP-​exo data. Front Genet 2014, 5:75.
26. Amemiya HM, Kundaje A, Boyle AP. The ENCODE Blacklist: Identification of
Problematic Regions of the Genome. Sci Rep 2019, 9(1):9354.
27. Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, Wong E, Orlov YL, Zhang W,
Jiang J et al. Integration of external signaling pathways with the core transcrip-
tional network in embryonic stem cells. Cell 2008, 133(6):1106–​1117.
28. Shen L, Shao NY, Liu X, Maze I, Feng J, Nestler EJ. diffReps: detecting dif-
ferential chromatin modification sites from ChIP-​seq data with biological
replicates. PLoS One 2013, 8(6):e65598.
29. Manser P, Reimers M. A simple scaling normalization for comparing ChIP-​Seq
samples. PeerJ PrePrints 2014, 1.
30. Tu S, Li M, Chen H, Tan F, Xu J, Waxman DJ, Zhang Y, Shao Z. MAnorm2
for quantitatively comparing groups of ChIP-​seq samples. Genome Res 2021,
31(1):131–​145.
31. Nair NU, Sahu AD, Bucher P, Moret BM. ChIPnorm: a statistical method for
normalizing and identifying differential regions in histone modification ChIP-​
seq libraries. PLoS One 2012, 7(8):e39573.
32. Taslim C, Wu J, Yan P, Singer G, Parvin J, Huang T, Lin S, Huang K. Comparative
study on ChIP-​seq data: normalization and binding pattern characterization.
Bioinformatics 2009, 25(18):2334–​2340.
33. Orlando DA, Chen MW, Brown VE, Solanki S, Choi YJ, Olson ER, Fritz CC,
Bradner JE, Guenther MG. Quantitative ChIP-​ Seq normalization reveals
global modulation of the epigenome. Cell Rep 2014, 9(3):1163–​1170.
34. Lun AT, Smyth GK. csaw: a Bioconductor package for differential binding ana-
lysis of ChIP-​seq data using sliding windows. Nucleic Acids Res 2016, 44(5):e45.
35. Zhang Y, Lin YH, Johnson TD, Rozek LS, Sartor MA. PePr: a peak-​calling pri-
oritization pipeline to identify consistent or differential peaks from replicated
ChIP-​Seq data. Bioinformatics 2014, 30(18):2568–​2575.
318 Next-Generation Sequencing Data Analysis

36. Xu H, Wei CL, Lin F, Sung WK. An HMM approach to genome-​wide iden-
tification of differential histone modification sites from ChIP-​ seq data.
Bioinformatics 2008, 24(20):2344–​2349.
37. Allhoff M, Sere K, J FP, Zenke M, I GC. Differential peak calling of ChIP-​seq
signals with replicates with THOR. Nucleic Acids Res 2016, 44(20):e153.
38. Stark R, Brown G. DiffBind: differential binding analysis of ChIP-​Seq peak
data. In R package version 2011, 100.
39. Liang K, Keles S. Detecting differential binding of transcription factors with
ChIP-​seq. Bioinformatics 2012, 28(1):121–​122.
40. Steinhauser S, Kurzawa N, Eils R, Herrmann C. A comprehensive comparison
of tools for differential ChIP-​seq analysis. Brief Bioinform 2016, 17(6):953–​966.
41. Eder T, Grebien F. Comprehensive assessment of differential ChIP-​seq tools
guides optimal algorithm selection. Genome Biol 2022, 23(1):119.
42. Chen L, Wang C, Qin ZS, Wu H. A novel statistical method for quantitative
comparison of multiple ChIP-​seq datasets. Bioinformatics 2015.
43. Taslim C, Huang T, Lin S. DIME: R-​ package for identifying differential
ChIP-​ seq based on an ensemble of mixture models. Bioinformatics 2011,
27(11):1569–​1570.
44. Schweikert G, Kuo D. MMDiff2: statistical testing for ChIP-​Seq data sets. In.,
vol. R package version 1.24.0; 2022.
45. Song Q, Smith AD. Identifying dispersed epigenomic domains from ChIP-​Seq
data. Bioinformatics 2011, 27(6):870–​871.
46. Yu G, Wang LG, He QY. ChIPseeker: an R/​ Bioconductor package for
ChIP peak annotation, comparison and visualization. Bioinformatics 2015,
31(14):2382–​2383.
47. McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, Wenger
AM, Bejerano G. GREAT improves functional interpretation of cis-​regulatory
regions. Nat Biotechnol 2010, 28(5):495–​501.
48. Zhu LJ, Gazin C, Lawson ND, Pages H, Lin SM, Lapointe DS, Green MR.
ChIPpeakAnno: a Bioconductor package to annotate ChIP-​seq and ChIP-​chip
data. BMC Bioinformatics 2010, 11:237.
49. Welch RP, Lee C, Imbriano PM, Patil S, Weymouth TE, Smith RA, Scott LJ,
Sartor MA. ChIP-​ Enrich: gene set enrichment testing for ChIP-​ seq data.
Nucleic Acids Res 2014.
50. Liu T, Ortiz JA, Taing L, Meyer CA, Lee B, Zhang Y, Shin H, Wong SS, Ma J, Lei
Y et al. Cistrome: an integrative platform for transcriptional regulation studies.
Genome Biol 2011, 12(8):R83.
51. Machanick P, Bailey TL. MEME-​ChIP: motif analysis of large DNA datasets.
Bioinformatics 2011, 27(12):1696–​1697.
52. Bailey TL, Johnson J, Grant CE, Noble WS. The MEME Suite. Nucleic Acids Res
2015, 43(W1):W39–​49.
53. Droit A, Gottardo R, Robertson G, Li L. rGADEM: de novo motif discovery. R
package version 2.44.0. . In.; 2022.
54. Thomas-​ Chollier M, Herrmann C, Defrance M, Sand O, Thieffry D, van
Helden J. RSAT peak-​motifs: motif analysis in full-​size ChIP-​seq datasets.
Nucleic Acids Res 2012, 40(4):e31.
55. Castro-​ Mondragon JA, Riudavets-​ Puig R, Rauluseviciute I, Lemma RB,
Turchi L, Blanc-​Mathieu R, Lucas J, Boddie P, Khan A, Manosalva Perez N
Mapping Protein-DNA Interactions with ChIP-Seq 319

et al. JASPAR 2022: the 9th release of the open-​access database of transcription
factor binding profiles. Nucleic Acids Res 2022, 50(D1):D165–​D173.
56. Mahony S, Benos PV. STAMP: a web tool for exploring DNA-​binding motif
similarities. Nucleic Acids Res 2007, 35(Web Server issue):W253–​258.
57. Gupta S, Stamatoyannopoulos JA, Bailey TL, Noble WS. Quantifying simi-
larity between motifs. Genome Biol 2007, 8(2):R24.
58. McLeay RC, Bailey TL. Motif Enrichment Analysis: a unified framework and
an evaluation on ChIP data. BMC Bioinformatics 2010, 11:165.
59. Bailey TL, Machanick P. Inferring direct DNA binding from ChIP-​seq. Nucleic
Acids Res 2012, 40(17):e128.
60. Bailey TL, Grant CE. SEA: Simple Enrichment Analysis of motifs. bioRxiv 2021,
doi: https://​doi.org/​10.1101/​2021.08.23.457​422.
61. Grant CE, Bailey TL, Noble WS. FIMO: scanning for occurrences of a given
motif. Bioinformatics 2011, 27(7):1017–​1018.
62. The MEME Suite (http://​meme.nbcr.net/​meme/​)
63. Ernst J, Kellis M. Discovery and characterization of chromatin states for sys-
tematic annotation of the human genome. Nat Biotechnol 2010, 28(8):817–​825.
64. Klein HU, Schafer M, Porse BT, Hasemann MS, Ickstadt K, Dugas M.
Integrative analysis of histone ChIP-​seq and transcription data using Bayesian
mixture models. Bioinformatics 2014, 30(8):1154–​1162.
65. Schafer M, Klein HU, Schwender H. Integrative analysis of multiple gen-
omic variables using a hierarchical Bayesian model. Bioinformatics 2017,
33(20):3220–​3227.
66. Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, Tang Q, Meyer CA, Zhang
Y, Liu XS. Target analysis by integration of transcriptome and ChIP-​seq data
with BETA. Nature protocols 2013, 8(12):2502–​2515.
67. Shin H, Liu T, Manrai AK, Liu XS. CEAS: cis-​regulatory element annotation
system. Bioinformatics 2009, 25(19):2605–​2606.
14 
Epigenomics by DNA Methylation
Sequencing

The genomic information embedded in the primary nucleotide sequence of


DNA is modulated by epigenomic code generated from chemical modifications
of DNA bases and key DNA-​interacting proteins such as the histones. The
methylation of cytosines leading to the formation of 5-​ methylcytosines
(5mCs), for example, provides a major means for the modification of the
primary DNA code. As detailed in Chapter 2, DNA methylation plays an
important role in many biological functions such as embryonic development,
cell differentiation, and stem cell pluripotency, through regulating gene
expression and chromatin remodeling. Abnormal patterns of DNA methyla-
tion, on the other hand, lead to diseases such as cancer. DNA methylome ana-
lysis, as a key component of epigenomics, has for many years been conducted
with the use of microarrays (such as the Illumina Infinium MethylationEPIC
BeadChips). While microarrays are low-​cost and easy to use, their inherent
constraints, such as limited genomic coverage from the use of pre-​selected
probes, and being available for only a few model organisms, have limited
their use. In comparison, NGS offers a more unbiased, comprehensive, and
quantitative approach for the study of DNA methylation status in a wide
array of species. This chapter focuses on DNA methylation sequencing data
generation and analysis. For epigenomic studies that involve interrogation of
histone modifications, ChIP-​seq (covered in Chapter 13) can be used.

14.1 DNA Methylation Sequencing Strategies


Because the DNA polymerases used in the regular NGS sequencing library
construction process cannot distinguish methylated from unmethylated
cytosines, DNA methylation pattern is usually not retained in the process. In
order to study DNA methylation status with NGS, different strategies need
to be used, including bisulfite conversion, methylated DNA enrichment,
and more recently enzymatic conversion-​based methyl-​seq. Bisulfite con-
version employs a chemical conversion process, which uses sodium bisulfite
to deaminate unmethylated cytosines. After the conversion, unmethylated

DOI: 10.1201/9780429329180-17 321


322 Next-Generation Sequencing Data Analysis

cytosines in a DNA molecule are converted to uracils, while 5mCs in the


same molecule are retained since they are non-​reactive. The subsequent
sequencing of the converted DNA, therefore, reads unmethylated cytosines
as thymines, and methylated cytosines as cytosines. The efficiency and spe-
cificity of this process can be monitored and optimized through the use of
certain methylated and unmethylated DNAs as controls. Based on genomic
coverage, this bisulfite sequencing approach can be further divided into
two subcategories as covered next. The more recently developed enzymatic
methyl-​seq is based on a similar approach, but the conversion of unmethylated
cytosines to uracils is based on enzymatic reactions, which is not DNA dam-
aging compared to the chemical method. The methylated DNA enrichment
approach relies on capturing of methylated DNA for targeted sequencing,
with the use of 5mC antibodies or proteins that bind to methylated cytosines.
This section details these major DNA methyl-​seq approaches.

14.1.1 Bisulfite Conversion Methyl-​Seq


14.1.1.1 Whole-​Genome Bisulfite Sequencing (WGBS)
As the name suggests, WGBS analyzes cytosine methylation in the entire
genome, i.e., the methylome. In preparing WGBS libraries from total gen-
omic DNA, regular DNA library construction protocols need to be modi-
fied. For example, if adaptors are added prior to the bisulfite conversion
step, they must not contain unmethylated cytosines, i.e., all cytosines in the
adaptor sequence must be methylated. In the PCR step, a polymerase that
can tolerate uracil residues needs to be used. As a result of the conversion
and subsequent PCR amplification, the two DNA strands that were originally
complementary are no longer complementary. Instead, four strands that are
distinct from the original complementary strands are generated (Figure 14.1).
Furthermore, the conversion leads to reduced sequence complexity due to
underrepresentation of cytosines in the generated reads. Without the use of
an external sequencing library to create a calibration table for basecaller, the
reduced sequence complexity will lead to high basecall error rate. Therefore,
use of a calibration library, such as the phiX174 control library for Illumina
sequencing, is needed for bisulfite sequencing data generation.
The power to detect DNA methylation levels, and differentially methylated
sites or regions between different experimental groups (e.g., disease vs.
normal), is dependent on sequencing depth and the number of biological
replicates in each group. In addition, key statistical factors, including mean
DNA methylation level at a given site, within-​group biological variation, and
between-​condition difference, also heavily affect detection power [1]. For
WGBS studies, a general guideline from the U.S. NIH Roadmap Epigenomics
Project recommends at least two replicates per condition with a combined
depth of at least 30× [2]. Consistent with this recommendation, Ziller and
colleagues suggested to use a sequencing depth of 5–​15× per WGBS sample,
Epigenomics by DNA Methylation Sequencing 323

Watson >>ACmGTTCGCTTGAG>> Cm methylated


Crick <<TGCmAAGCGAACTC<< C Un-methylated

1) Denaturation

Watson >>ACmGTTCGCTTGAG>> Crick <<TGCmAAGCGAACTC<<

2) Bisulfite Treatment

BSW >>ACmGTTUGUTTGAG>> BSC <<TGCmAAGUGAAUTU<<

3) PCR Amplification

BSW >>ACmGTTTGTTTGAG>> BSC <<TGCmAAGTGAATTT<<


BSWR <<TG CAAACAAACTC<< BSCR >>ACG TTCACTTAAA>>

FIGURE 14.1
Major steps of bisulfite sequencing. Prior to bisulfite treatment, the two strands of DNA are
first separated by denaturation. The bisulfite treatment then converts unmethylated, but not
methylated, cytosines to uracils. The two strands from the treatment, BSW and BSC, are then
subjected to PCR amplification. This leads to the generation of four strands (BSW, BSWR, BSC,
and BSCR), all of which are distinct from the original Watson and Crick strands. (From Xi Y. and
Li W. (2009) BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinformatics,
10, 232. Used under the terms of the Creative Commons Attribution License (http://​crea​tive​
comm​ons.org/​licen​ses/​by/​2.0). © 2009 Xi and Li.)

and that sequencing at higher depth may not be as cost effective as adding
more biological replicates in reaching higher detection power [3].

14.1.1.2 Reduced Representation Bisulfite Sequencing (RRBS)


While WGBS enables detection of methylation in the entire genome, the cost
associated with such analyses is relatively high. To reduce the cost, strat-
egies such as RRBS [4] were devised. To perform RRBS, genomic DNA is first
digested with a methylation-​insensitive restriction enzyme (such as MspI)
that recognizes CpG-​containing restriction site. The digested DNA products
are then separated and size selected to pick fragments in a certain size range
for bisulfite conversion and then sequencing. While it has limited coverage
of CpG-​poor regions of the genome, RRBS provides a rough survey of DNA
methylation by examining 4–​17% of the 28 million CpG dinucleotides in
the human genome [5]. If particular region(s) of the genome are found to
be of special interest, they can be captured for subsequent sequencing using
324 Next-Generation Sequencing Data Analysis

approaches such as ligation capture [6, 7], bisulfite padlock probes [8], or
liquid hybridization capture [9, 10].

14.1.2 Enzymatic Conversion Methyl-​Seq


Besides the bisulfite-​based chemical method, the conversion of cytosine to
uracil through deamination can also be achieved using enzymes such as
APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide-​
like). Besides unmethylated cytosine, however, APOBEC can also deaminate
5mC and 5hmC (Chapter 2). To protect them from deamination in order
to quantify their presence, two other enzymes TET2 (Tet methylcytosine
dioxygenase 2) and BGT (β-​glucosyltransferase) need to be used first to con-
vert 5mC to 5caC, and 5hmC to 5-​ghmC (5-​glucosylhydroxymethylcytosine),
respectively. After such enzymatic modifications, 5caC and 5-​ghmC are no
longer substrates of APOBEC. In the subsequent APOBEC-​based deamin-
ation step, only unmethylated cytosines are converted to uracils, allowing
determination of 5mCs and 5hmCs in the original sample. Compared to
bisulfite treatment, enzyme-​based conversion does not cause DNA damage
and sample loss. This leads to increased detection accuracy, sensitivity, and
coverage of CpGs and high GC content regions [11–​13]. The increased sen-
sitivity makes it possible to study single cells, cell-​free DNA, or FFPE DNA.
With enzymatic conversion, long-​range methylation information can also be
retained and studied using long-read NGS technologies [14].

14.1.3 Enrichment-​Based Methyl-​Seq
Different from the above bisulfite conversion-​based methods, the methylated
DNA enrichment strategy captures methylated DNA for targeted sequencing.
One of the methods based on this strategy is MeDIP-​seq, or methylated DNA
immunoprecipitation coupled with NGS. In this method, antibodies against
5mC are used to precipitate methylated single-​stranded DNA fragments for
sequencing. Another commonly used method is MBD-​seq, or methyl-​CpG-​
binding domain capture (MBDCap) followed by NGS. MBD-​seq utilizes
proteins such as MBD2 or MECP2 that contain the methyl-​CpG-binding
domain to enrich for methylated double-​stranded DNA fragments. In one
type of MBDCap method called MIRA (Methylated-​CpG Island Recovery
Assay), a protein complex of MBD2 and MBD3L1 (methyl-​CpG-​binding
domain protein 3-​like-​1) is used to achieve enhanced affinity to methylated
CpG regions. While MeDIP-​seq and MBD-​aeq usually generate highly con-
cordant results, there are some differences between these two approaches.
MeDIP-​seq can detect both CpG and non-​CpG methylation, while MBD-​seq
is focused on methylated CpG sites because of the binding affinity of MBD.
At methylated CpG sites, MeDIP tends to enrich at regions that have low
CpG density, while MBD-​seq favors regions of relatively higher CpG content
[15, 16].
Epigenomics by DNA Methylation Sequencing 325

In principle, these enrichment-​based methods are very similar to ChIP-​


seq (Chapter 13), based on the same process of specific protein-​based DNA
capture, protein-​DNA complex affinity binding, and target DNA elution.
Likewise, their sequencing data generation and subsequent analysis are also
similar to those in ChIP-​seq. Therefore, the data analysis methods covered
in Chapter 13 equally apply to the analysis of sequencing data generated
by MeDIP-​seq, MBD-​seq, or other methylated DNA enrichment-​based NGS
methods. This chapter is, therefore, mostly focused on the analysis of chem-
ical or enzymatic conversion-​based methyl-​seq data.

14.1.4 Differentiation of Cytosine Methylation from


Demethylation Products
Besides 5mC, cytosine demethylation products, such as 5hmC (Figure 14.2),
have also received growing interest because of their potential role as a new
epigenetic marker [17]. Although the regular enzymatic conversion-​based
methyl-​seq procedure detects both 5mC and 5hmC indiscriminately, a modi-
fied version can be used to detect 5hmC alone, in which the step of TET2 pro-
tection is skipped leaving 5mC unprotected while the glucosylation of 5hmC
by BGT still protects 5-​hmC from deamination by APOBEC. By combining
results from the regular and the modified versions of the enzymatic methyl-​
seq, separate detection of both 5mC and 5hmC is achieved.
Other bisulfite conversion-​based approaches to distinguish 5hmC from
5mC include TAB-​ seq (or TET-​ assisted bisulfite sequencing) [18] and
oxBS-​seq (or oxidative bisulfite sequencing) [19]. Among the three 5mC
demethylation intermediate products (5hmC, 5fC, and 5caC, see Chapter 2),
5fC and 5caC are converted to uracils by sodium bisulfite but 5hmC (like
5mC) is not. During subsequent sequencing, as a consequence, 5hmC cannot
be differentiated from 5mC, while 5fC/​5caC cannot be differentiated from
unmethylated cytosine. In the instance of oxBS-​seq, 5hmC is first oxidized to
5fC before the bisulfite conversion step. Since the 5fC is then converted into
uracil, subsequent sequencing and analysis provides information on 5mC

Cytosine (C) 5-Methylcytosine 5-Hydroxymethylcytosine


(5mC) (5hmC)

NH2

HO N

N O
H
FIGURE 14.2
The chemical structures of cytosine, 5-​methylcytosine (5mC), and 5-​hydroxymethylcytosine
(5hmC).
326 Next-Generation Sequencing Data Analysis

alone. After subtracting oxBS-​seq result from regular bisulfite sequencing


result, information on 5hmC is revealed.
Differentiation of 5fC and 5caC has also been made possible with recent
method development [20, 21], but their levels are found to be typically low.
Some third-​generation single-​molecule sequencing technologies, such as the
Pacific Biosciences’ SMRT sequencing and nanopore sequencing, have been
shown to be capable of differentially detecting these different modifications
without relying on bisulfite or enzymatic conversion [22–​26]. The SMRT plat-
form detects various base modifications based on polymerase dynamics as
reflected in the pulses of light emitted from incorporation of nucleotides,
while the nanopore platform is based on recognition of specific patterns in
the ionic current signal.

14.2 DNA Methylation Sequencing Data Analysis


14.2.1 Quality Control and Preprocessing
After raw data generation, the quality control (QC) step (Figure 14.3)
removes low-​quality reads or basecalls as they directly affect subsequent
alignment to the reference genome and DNA methylation site identification.
The general data QC steps detailed in Chapter 5 should be performed for
their removal. Other QC steps include adapter trimming as some sequen-
cing reactions may run through DNA inserts into adapters. In addition, for
MspI-​digested RRBS libraries, the DNA fragment end repair step during
the library construction artificially introduces two bases (an unmethylated
cytosine and a guanine) to both ends, both of which should be trimmed
off as well. Tools such as Trim Galore (a wrapper tool using Cutadapt and
FastQC) [27] can be used for these trimming steps, especially removing the
two artificially introduced bases in RRBS reads derived from MspI diges-
tion. Besides these general-​purpose QC tools, some packages designed for
bisulfite sequencing reads processing, including BSmooth [28] and WBSA
[29], also contain QC modules.

14.2.2 Read Mapping
In order to identify methylated DNA sites, sequencing reads derived from
bisulfite or enzymatic conversion, or methylated DNA enrichment, need to
be first mapped to the reference genome. Mapping of reads generated from
the enrichment-​based methods is rather straightforward, and like mapping
ChIP-​seq reads, is usually conducted with general aligners, such as Bowtie,
BWA, or SOAP. Mapping of bisulfite or enzymatic conversion-​based methyl-​
seq reads, however, is less straightforward. This is because through the
Epigenomics by DNA Methylation Sequencing 327

DNA Methyl-Seq Data


QC &Preprocessing

Read Mapping

Post Mapping QC

Methylation/Demethylation
Level Quantification

Visualization

Differential Methylation
Analysis

Validation and Interpretation

FIGURE 14.3
Major steps of chemical or enzymatic conversion-​based DNA methyl-​seq data analysis.

bisulfite or enzymatic conversion and the subsequent sequencing process,


a converted unmethylated cytosine is read as a thymine (T), or an adenine
(A) on the complementary strand, while a methylated cytosine remains as a
cytosine (C) or a guanine (G) on the complementary strand (see Figure 14.1).
Such base changes have several implications for the read mapping process:

• Fuzziness in mapping: A T in the reads could be mapped to a C or T in the


reference sequence, thus complicating the searching process.
• Increase in search space: This is partly caused by the non one-​to-​one
mapping, and more seriously, by the generation of the four bisulfite-​
converted strands that are distinct from the reference strands (also
illustrated in Figure 14.1), leading to significant increase in search space.
• Reduction in sequence complexity: The amount of Cs in the bisulfite reads
is significantly reduced, and this reduction in sequence complexity
leads to higher levels of mapping ambiguity. Consequently, aligning
bisulfite sequencing reads to the reference genome is not as straightfor-
ward as that for ChIP-​seq or other DNA deep sequencing data.
328 Next-Generation Sequencing Data Analysis

Without making modifications to the typical DNA alignment process, the


methylation status of a cytosine in the genome will affect alignment of
reads covering the region. There are two general strategies for mapping
conversion-​based methylation sequencing reads: (1) replacing all Cs in the
reference genome with the wild-​card letter Y to match both Cs and Ts in the
reads; (2) converting all Cs in the reference sequence and reads to Ts, and
then aligning with a seed-​and-​extend approach (see Table 14.1). Aligners that
use the wild-​card approach include BSMAP [30], Last [31], GSNAP [32], and
RRBSMAP (a version of BSMAP specifically tailored for RRBS reads, merged
into BSMAP-​2.0) [33]. In the example of BSMAP, it uses SOAP for carrying
out read alignment, and deploys genome hashing and bitwise masking for
speed and accuracy. BSMAP indexes the reference genome using hash table
containing original reference seed sequences and all their possible bisulfite
conversion variants through the replacement of Cs with Ts. After determining
the potential genomic position of each read by looking up the hash
table, for the T(s) in each bisulfite read that are mapped to reference genome
position(s) where the original reference base(s) are C(s), BSMAP masks as

TABLE 14.1
Read Mapping Tools for Chemical or Enzymatic Conversion-​Based DNA Methylation
Sequencing
Name Description Reference

Three-​Letter Aligners
Bismark Deploys Bowtie 2 (or HISAT2) for alignment, and performs [34]
cytosine methylation calling
bwa-​meth Wraps BWA-​MEM, provides local alignment for speed and [35]
accurary even without trimming
BS-​Seeker2/​ Incorporates major aligners, such as Bowtie 2, to achieve [36, 37]
BS-​Seeker3 gapped local alignment. BS-​Seeker3 further improves speed
and accuracy, and offers post-​alignment analysis including
QC and visualization
BSmooth Uses Bowtie 2 (or Merman) for alignment. Also provides QC, [28]
smoothing-​based methylation quantification, and differential
methylation detection
Wild-​Card Aligners
BSMAP/​ Combines hash table seeding incorporated in SOAP, and [30]
RRBSMAP bitwise masking to achieve speed and accuracy
GSNAP Employs hash tables built for plus and minus strands using C-​ [32]
T/​G-​A substitutions
Last Builds on the traditional alignment strategy of seed-​and-​ [31]
extend (like Blast), but with use of adaptive seeds
BatMeth2 Performs indel-​sensitive mapping, DNA methylation [41]
quantification, differential methylation detection, annotation,
and visualization
Epigenomics by DNA Methylation Sequencing 329

C(s). The masked bisulfite reads are then mapped again to the reference
genome.
Aligners such as Bismark [34], bwa-​meth [35], and BS-​Seeker2/​BS-​Seeker3
[36, 37] use the other (three-​letter) approach. One advantage of this approach
is that fast mapping algorithms such as BWA-​MEM or bowtie2 can be used.
For example, Bismark carries out alignment by first converting Cs in the
reads into Ts, and Gs into As (equivalent of the C-​to-​T conversion on the com-
plementary strand) (Figure 14.4). This conversion process is also performed
on the reference genome. The converted reads are then aligned, using Bowtie
or Bowtie2, to the converted reference genome in four parallel processes (also
refer to Figure 14.1), out of which a unique best alignment is determined
[alignment (1) in Figure 14.4]. Among the above wild-​card and three-​letter
methods, benchmark studies [38–​40] found that bwa-​meth, Bismark, and
BSMAP offer a good combination of accuracy, speed, and genomic region
coverage.

Genomic fragment
sequence after bisulfite
treatment

Read conversion

Align to bisulfite
converted genomes

Forward strand C-to-T converted genome Forward strand G-toA converted genome

Read all four alignment


outputs simultaneously
to determine if the
sequence can be
mapped uniquely
Determine unqiue best alignment
FIGURE 14.4
The “three-​letter” bisulfite sequencing read alignment approach used by Bismark. (Adapted
from Krueger F. and Andrews S.R. (2011) Bismark: a flexible aligner and methylation caller
for Bisulfite-​Seq applications. Bioinformatics, 27, 1571–​2. Used under the terms of the Creative
Commons Attribution Non-​ Commercial License (http://​crea​tive​comm​ons.org/​licen​ses/​by-​
nc/​2.5). © 2011 Krueger and Andrews.)
330 Next-Generation Sequencing Data Analysis

After mapping, additional QC is needed prior to extraction of methylation


information. For example, reads that have low MAPQ (e.g., < 10) should be
removed. Duplicate reads that map exactly to the same position are most
likely PCR artifacts and should be removed from further analysis (except in
the case of RRBS). Methylation bias (or mbias) should also be checked and
corrected. This bias is caused by the step of end repair during library prep
post fragmentation, which incorporates unmethylated Cs at both ends of
repaired DNA fragments leading to artificially low methylation levels at the
affected sites [42]. Mbias plot, showing mean methylation levels at all base
positions across the entire length of sequence reads, can be used to reveal
the magnitude of the bias, based on which affected base positions should be
excluded from further analysis. In addition, polymorphic sites which involve
C→T variation (G→A on the complementary strand) will also affect analysis
and should also be excluded. As part of post-​mapping QC, distribution of the
mapped reads in the genome should be visually examined. This provides an
initial survey of the results, and at the same time may reveal other abnormal-
ities such as significantly unbalanced numbers of reads mapped to the two
DNA strands in a genomic region (this should be inspected with caution and
the reads may need to be filtered out). Methyl-​seq QC tools such as BSeQC
[43], and post-​mapping analysis tools such as MethylDackel [44], can be used
to carry out these post-​mapping QC steps.

14.2.3 Quantification of DNA Methylation/​Demethylation Products


After read mapping, uniquely mapped reads need to be aggregated to quan-
tify the methylation level (also called β-​value) at individual cytosine sites
in the reference genome, based on the frequency of Cs (i.e. methylated
cytosines) and Ts (unmethylated cytosines) in reads mapped to each of these
sites. This quantitative step can be performed by dividing the total number of
Cs by the total combined number of Cs and Ts that are mapped to each site.
All of the sequence mappers introduced in the previous section generate this
information. MethylDackel and other post-​mapping tools such as methylKit
[45] can also be used for methylation quantification. If separate quantification
of 5mC and other demethylation products (such as 5hmC) is needed after
applying approaches such as oxBS-​seq, TAB-​seq, or enzymatic methyl-​seq,
simultaneous estimation of methylation/​demethylation product levels can
be achieved using methods such as MLML [46], which is part of the MethPipe
package [47]. For this quantification step, it should be noted that the involved
calculations usually require a minimum depth, e.g., at least three reads, at
the individual sites to avoid deriving unreliable methylation levels from too
few reads.
Besides quantifying methylation levels at individual cytosine sites, DNA
methylation quantification is also calculated on a region-​by-​region basis,
usually performed to facilitate comparisons between multiple samples.
Epigenomics by DNA Methylation Sequencing 331

Different approaches can be used for regional DNA methylation quantifi-


cation. These include approaches to segment the genome into bins, sliding
windows of user-​defined size (e.g. 100 bp) [48], or predefined regions (such
as promoters, CpG islands, gene bodies, introns, etc.). The mean of methyla-
tion levels of individual cytosine sites, or alternatively the overall proportion
of methylated cytosines among all cytosines, within each of such regions can
be used to represent each region.
These calculations, however, do not take into consideration the possible
existence of SNPs that involve the change from C to T. Some algorithms,
such as Bis-​ SNP [49], remove this potential confounding factor through
distinguishing conversion-​caused base changes from genetic variants. The
use of sequence reads from the complementary strand makes this possible,
because a T produced from bisulfite conversion will have a G on the opposite
strand while a C→T SNP will have an A on the other strand.
Different from the conversion-​based sequencing methods, the methylated
DNA enrichment sequencing approaches such as MeDIP-​seq and MBD-​seq
cannot quantify methylation at the single-​ nucleotide resolution. In add-
ition, the absolute levels of DNA methylation cannot be obtained from the
enrichment-​based methods, as the sequence read counts from these methods
are a function of both absolute DNA methylation levels and regional CpG
content. Since these approaches are based on affinity immunoprecipitation
and more similar to ChIP-​seq, analytical methods developed for ChIP-​seq
data analysis, including background determination, normalization, and peak
detection, can be applied for quantification of DNA methylation by these
approaches. As an output, the degree of DNA methylation can be summarized
as coverage over a predefined region, such as per gene, promoter, or certain
sized bin.

14.2.4 Visualization
Visualizing DNA methylation data serves at least two purposes. Firstly, distri-
bution pattern of DNA methylation may be discerned through visualization.
Secondly, visual examination of known DNA methylation regions and other
randomly selected regions also offers data validation and a quick estimate of
data quality. One method to visualize DNA methyl-​seq data and associated
information, such as depth of coverage, is through the use of bedGraph files.
This standard format (Figure 14.5), compatible with most genome browsers
and tools including the Washington University EpiGenome Browser [50],
can be directly generated from many of the methylation quantification tools
such as Bismark and methylKit. Figure 14.6 shows an example of displaying
methylation level along with read depth in the genome.
Alternatively, DNA methylation quantification results can be saved in tab-​
delimited files and then converted to bigBed or bigWig formats [51]. Both
formats are compatible with and enable visualization of the methylation
332 Next-Generation Sequencing Data Analysis

track type=bedGraph
chr19 45408804 45408805 1.0
chr19 45408806 45408807 0.75
chr19 45408854 45408855 0.3
chr19 45408855 45408856 0.5
FIGURE 14.5
An example of the bedGraph file format. It includes a track definition line (the first line),
followed by track data lines in four column format, i.e. chromosome, chromosome start position,
chromosome end position, and data value.

results in web-​based genome browsers such as the UCSC Genome Browser,


or desktop-​based ones such as IGV or IGB. An additional option is to export
DNA methylation data to the VCF format using tools such as GobyWeb [52],
and then visualize with genome browsers such as IGV or Savant.

14.3 Detection of Differentially Methylated


Cytosines and Regions
One frequent goal of DNA methylation analysis is to compare and identify
specific cytosines and genomic regions that show differential methylation
between conditions. To identify differentially methylated cytosines
and regions (DMCs/​DMRs), different tools employing different statistical
methods have been developed (see Table 14.2). The statistical methods
employed by these tools include parametric tests such as t-​test or ANOVA,
and nonparametric tests such as Fisher’s exact test, Wilcoxon test, Chi-​square
test, or Kruskal–​Wallis test. The parametric tests assume normal distribution,
which is likely to be violated for DNA methylation data as it tends to follow
bimodal distribution. As a result, most currently available tools use non-
parametric tests. For example, methylKit and RnBeads [53, 54] use Fisher’s
exact test for comparison of groups without replicates. This test can also be
applied directly often with good performance. For comparisons involving
multiple samples per group, methylKit and RnBeads use logistic and
linear regression, respectively. WBSA employs Wilcoxon test, and BSmooth
applies a modified t-​test with local data smoothing to increase detection
power. Another package called Methy-​Pipe [55] detects DMRs using the
Mann-​Whitney U test with a sliding window approach. More sophisticated
approaches include the use of a beta-​binomial hierarchical model in MOABS
[56], and Shannon entropy in QDMR [57]. Besides these different statistical
tests or models, another notable difference among these tools is on how bio-
logical replicates are handled. Earlier methods tend to pool replicate data
for DMC/​DMR detection, leading to the loss of information on sample-​to-​
sample variation. Newer methods, such as BSmooth and MOABS, are more
newgenrtpdf
Epigenomics by DNA Methylation Sequencing
FIGURE 14.6
Visualization of DNA methylation data in a genome browser. Shown here is the methylC track in the Washington University EpiGenome
Browser, for a region of the human chromosome 7 where the HOXA gene cluster is located. The original WGBS data was collected from H1
human embryonic stem cells. Both DNA methylation levels (represented by vertical bars) and read depth (the smoothed black curve) are
displayed in a strand-​specific fashion. The different foreground/​background colors of the vertical bars represent different cytosine contexts,
with CG represented by blue/​grey, CHG by orange/​light orange, and CHH by magenta/​light magenta. A zoomed-​in view of the red boxed
region is shown on the top. The left axis marks DNA methylation level, while the right marks read depth. (From Zhou X. et al. (2014) methylC
Track: visual integration of single-​base resolution DNA methylation data on the WashU EpiGenome Browser. Bioinformatics, 30, 2206–​7.
Modified and used with permission from Oxford University Press. © 2014 Zhou et al.)

333
334 Next-Generation Sequencing Data Analysis

TABLE 14.2
Tools for Detection of Differentially Methylated Cytosines/​Regions
Name Description Reference

Fisher’s Applies directly the classical Fisher’s exact test for DMC/​DMR N/​A
recognition
methylKit Uses logistic regression for groups with replicates, and Fisher’s [45]
exact test if without replicates
methylSig Identifies DMCs/​DMRs using likelihood ratio estimation based [61]
on a beta-​binomial distribution model
RnBeads Combines statistical testing p-values, and priority ranking based [53, 54]
on absolute and relative effect size
RADMeth Uses log-​likelihood ratio test based on a beta-​binomial regression [62]
model for differential methylation testing
DMRFinder Applies Wald and empirical Bayes tests for differential [63]
methylation detection based on beta-​binomial modelling
Metilene A nonparametric method based on segmentation of the genome [64]
using a circular binary segmentation algorithm
DSS Uses a Bayesian hierarchical model to allow information sharing [65]
across different CpG sites, and Wald test for DMC detection

replicate-​
aware and provide estimation on biological variation, thereby
increasing detection power. On multiple testing correction, FDR is mostly
used, while other methods are also reported, such as a sliding linear model
(SLIM) method used by methylKit. Among the currently available DMC/​
DMR detection tools, benchmarking studies show that methylKit, Fisher’s
exact test, methylSig, and DMRFinder are among the top performers [58, 59].
Data obtained from methylated DNA enrichment-​ based approaches
follows the negative binomial distribution, like the ChIP-​seq and RNA-​seq
data. Therefore, they can be analyzed to identify DMRs using algorithms
developed for RNA-​seq-​based differential expression. For example, tools
such as EdgeR and DESeq can be directly used. In some DNA methylation
analysis tools, such as Repitools [60], EdgeR is directly called.

14.4 Data Verification, Validation, and Interpretation


The DMCs/​DMRs identified in the previous step need to be verified and fur-
ther validated. Verification is usually conducted on the same set of samples
as those used for DNA methylation sequencing data generation. Further
validation, on the other hand, is carried out on a new set of samples. For
DNA methylation sequencing data verification and validation, the following
Epigenomics by DNA Methylation Sequencing 335

techniques are often used: methylation-​specific PCR (such as MethyLight),


or methylation-​independent PCR coupled with pyrosequencing, mass spec-
trometry, or combined bisulfite restriction analysis (COBRA).
Data interpretation is a key step to translate a list of DMCs/​DMRs into
mechanistic understanding of the biological process under study. Most
potential effects of the DMCs/​DMRs can only be revealed through examining
them in their genomic context. Tools such as EpiExplorer [66], methylKit, or
WBSA can be very helpful in this regard via placing them in the context of
other genomic features such as CpG islands, transcription start sites, histone
modification marks, or repetitive regions. DMCs/​DMRs can also be mapped
to nearby genes, which can then be subjected to gene set enrichment, bio-
logical pathway, and gene networking analyses. In this regard, the web-​based
Genomic Regions Enrichment of Annotations Tool (or GREAT) [67] can be
used to map DMCs/​DMRs to nearby genes, while controlling for gene size
difference and distance, for functional annotation and interpretation.

References
1. Seiler Vellame D, Castanho I, Dahir A, Mill J, Hannon E. Characterizing the
properties of bisulfite sequencing data: maximizing power and sensitivity to
identify between-​group differences in DNA methylation. BMC Genomics 2021,
22(1):446.
2. Standards and Guidelines for Whole Genome Shotgun Bisulfite Sequencing
(www.roa​dmap​epig​enom​ics.org/​protoc​ols)
3. Ziller MJ, Hansen KD, Meissner A, Aryee MJ. Coverage recommendations
for methylation analysis by whole-​genome bisulfite sequencing. Nat Methods
2015, 12(3):230–​232.
4. Meissner A, Gnirke A, Bell GW, Ramsahoye B, Lander ES, Jaenisch R. Reduced
representation bisulfite sequencing for comparative high-​ resolution DNA
methylation analysis. Nucleic Acids Res 2005, 33(18):5868–​5877.
5. Sun Z, Cunningham J, Slager S, Kocher JP. Base resolution methylome pro-
filing: considerations in platform selection, data preprocessing and analysis.
Epigenomics 2015, 7(5):813–​828.
6. Nautiyal S, Carlton VE, Lu Y, Ireland JS, Flaucher D, Moorhead M, Gray JW,
Spellman P, Mindrinos M, Berg P et al. High-​throughput method for analyzing
methylation of CpGs in targeted genomic regions. Proc Natl Acad Sci U S A
2010, 107(28):12587–​12592.
7. Varley KE, Mitra RD. Bisulfite Patch PCR enables multiplexed sequen-
cing of promoter methylation across cancer samples. Genome Res 2010,
20(9):1279–​1287.
8. Deng J, Shoemaker R, Xie B, Gore A, LeProust EM, Antosiewicz-​Bourget J,
Egli D, Maherali N, Park IH, Yu J et al. Targeted bisulfite sequencing reveals
changes in DNA methylation associated with nuclear reprogramming. Nat
Biotechnol 2009, 27(4):353–​360.
336 Next-Generation Sequencing Data Analysis

9. Ivanov M, Kals M, Kacevska M, Metspalu A, Ingelman-​Sundberg M, Milani


L. In-​solution hybrid capture of bisulfite-​converted DNA for targeted bisulfite
sequencing of 174 ADME genes. Nucleic Acids Res 2013, 41(6):e72.
10. Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV, Consortium C.
Sensitive and specific multi-​cancer detection and localization using methyla-
tion signatures in cell-​free DNA. Ann Oncol 2020, 31(6):745–​759.
11. Han Y, Zheleznyakova GY, Marincevic-​ Zuniga Y, Kakhki MP, Raine A,
Needhamsen M, Jagodic M. Comparison of EM-​seq and PBAT methylome
library methods for low-​input DNA. Epigenetics 2021, 17(10):1195–​1204.
12. Vaisvila R, Ponnaluri VKC, Sun Z, Langhorst BW, Saleh L, Guan S, Dai N,
Campbell MA, Sexton BS, Marks K et al. Enzymatic methyl sequencing detects
DNA methylation at single-​base resolution from picograms of DNA. Genome
Res 2021, 31(7):1280–​1289.
13. Feng S, Zhong Z, Wang M, Jacobsen SE. Efficient and accurate determination
of genome-​ wide DNA methylation patterns in Arabidopsis thaliana with
enzymatic methyl sequencing. Epigenetics Chromatin 2020, 13(1):42.
14. Sun Z, Vaisvila R, Hussong LM, Yan B, Baum C, Saleh L, Samaranayake M,
Guan S, Dai N, Correa IR, Jr. et al. Nondestructive enzymatic deamination
enables single-​molecule long-​read amplicon sequencing for the determination
of 5-​methylcytosine and 5-​hydroxymethylcytosine at single-​base resolution.
Genome Res 2021, 31(2):291–​300.
15. Harris RA, Wang T, Coarfa C, Nagarajan RP, Hong C, Downey SL, Johnson BE,
Fouse SD, Delaney A, Zhao Y et al. Comparison of sequencing-​based methods
to profile DNA methylation and identification of monoallelic epigenetic
modifications. Nat Biotechnol 2010, 28(10):1097–​1105.
16. Nair SS, Coolen MW, Stirzaker C, Song JZ, Statham AL, Strbenac D, Robinson
MD, Clark SJ. Comparison of methyl-​DNA immunoprecipitation (MeDIP)
and methyl-​CpG binding domain (MBD) protein capture for genome-​wide
DNA methylation analysis reveal CpG sequence coverage bias. Epigenetics
2011, 6(1):34–​44.
17. Rodriguez-​Aguilera JR, Ecsedi S, Goldsmith C, Cros MP, Dominguez-​Lopez
M, Guerrero-​Celis N, Perez-​Cabeza de Vaca R, Chemin I, Recillas-​Targa F,
Chagoya de Sanchez V et al. Genome-​wide 5-​hydroxymethylcytosine (5hmC)
emerges at early stage of in vitro differentiation of a putative hepatocyte pro-
genitor. Sci Rep 2020, 10(1):7822.
18. Yu M, Han D, Hon GC, He C. Tet-​Assisted Bisulfite Sequencing (TAB-​seq).
Methods Mol Biol 2018, 1708 :645–​663.
19. Booth MJ, Branco MR, Ficz G, Oxley D, Krueger F, Reik W, Balasubramanian
S. Quantitative sequencing of 5-​methylcytosine and 5-​hydroxymethylcytosine
at single-​base resolution. Science 2012, 336(6083):934–​937.
20. Booth MJ, Marsico G, Bachman M, Beraldi D, Balasubramanian S. Quantitative
sequencing of 5-​formylcytosine in DNA at single-​base resolution. Nat Chem
2014, 6(5):435–​440.
21. Liu Y, Hu Z, Cheng J, Siejka-​Zielinska P, Chen J, Inoue M, Ahmed AA,
Song CX. Subtraction-​ free and bisulfite-​free specific sequencing of 5-​
methylcytosine and its oxidized derivatives at base resolution. Nat Commun
2021, 12(1):618.
Epigenomics by DNA Methylation Sequencing 337

22. Flusberg BA, Webster DR, Lee JH, Travers KJ, Olivares EC, Clark TA, Korlach
J, Turner SW. Direct detection of DNA methylation during single-​molecule,
real-​time sequencing. Nat Methods 2010, 7(6):461–​465.
23. Laszlo AH, Derrington IM, Brinkerhoff H, Langford KW, Nova IC, Samson
JM, Bartlett JJ, Pavlenok M, Gundlach JH. Detection and mapping of 5-​
methylcytosine and 5-​hydroxymethylcytosine with nanopore MspA. Proc Natl
Acad Sci U S A 2013, 110(47):18904–​18909.
24. Schreiber J, Wescoe ZL, Abu-​Shumays R, Vivian JT, Baatar B, Karplus K, Akeson
M. Error rates for nanopore discrimination among cytosine, methylcytosine,
and hydroxymethylcytosine along individual DNA strands. Proc Natl Acad Sci
U S A 2013, 110(47):18910–​18915.
25. Tse OYO, Jiang P, Cheng SH, Peng W, Shang H, Wong J, Chan SL, Poon LCY,
Leung TY, Chan KCA et al. Genome-​ wide detection of cytosine methyla-
tion by single molecule real-​time sequencing. Proc Natl Acad Sci U S A 2021,
118(5):e2019768118.
26. Rand AC, Jain M, Eizenga JM, Musselman-​Brown A, Olsen HE, Akeson M,
Paten B. Mapping DNA methylation with high-​throughput nanopore sequen-
cing. Nat Methods 2017, 14(4):411–​413.
27. Trim Galore! (www.bio​info​rmat​ics.babra​ham.ac.uk/​proje​cts/​trim​_​gal​ore/​)
28. Hansen KD, Langmead B, Irizarry RA. BSmooth: from whole genome bisulfite
sequencing reads to differentially methylated regions. Genome Biol 2012,
13(10):R83.
29. Liang F, Tang B, Wang Y, Wang J, Yu C, Chen X, Zhu J, Yan J, Zhao W, Li
R. WBSA: web service for bisulfite sequencing data analysis. PLoS One 2014,
9(1):e86707.
30. Xi Y, Li W. BSMAP: whole genome bisulfite sequence MAPping program.
BMC Bioinformatics 2009, 10:232.
31. Frith MC, Mori R, Asai K. A mostly traditional approach improves alignment
of bisulfite-​converted DNA. Nucleic Acids Res 2012, 40(13):e100.
32. Wu TD, Nacu S. Fast and SNP-​tolerant detection of complex variants and spli-
cing in short reads. Bioinformatics 2010, 26(7):873–​881.
33. Xi Y, Bock C, Muller F, Sun D, Meissner A, Li W. RRBSMAP: a fast, accurate
and user-​friendly alignment tool for reduced representation bisulfite sequen-
cing. Bioinformatics 2012, 28(3):430–​432.
34. Krueger F, Andrews SR. Bismark: a flexible aligner and methylation caller for
Bisulfite-​Seq applications. Bioinformatics 2011, 27(11):1571–​1572.
35. Pedersen BS, Eyring K, De S, Yang IV, Schwartz DA. Fast and accurate
alignment of long bisulfite-​seq reads. arXiv preprint arXiv:14011129 2014.
36. Huang KYY, Huang YJ, Chen PY. BS-​Seeker3: ultrafast pipeline for bisulfite
sequencing. BMC Bioinformatics 2018, 19(1):111.
37. Guo W, Fiziev P, Yan W, Cokus S, Sun X, Zhang MQ, Chen PY, Pellegrini M.
BS-​Seeker2: a versatile aligning pipeline for bisulfite sequencing data. BMC
Genomics 2013, 14 :774.
38. Kunde-​Ramamoorthy G, Coarfa C, Laritsky E, Kessler NJ, Harris RA, Xu M,
Chen R, Shen L, Milosavljevic A, Waterland RA. Comparison and quantitative
verification of mapping algorithms for whole-​genome bisulfite sequencing.
Nucleic Acids Res 2014, 42(6):e43.
338 Next-Generation Sequencing Data Analysis

39. Nunn A, Otto C, Stadler PF, Langenberger D. Comprehensive benchmarking


of software for mapping whole genome bisulfite data: from read alignment to
DNA methylation analysis. Brief Bioinform 2021, 22(5):bbab021.
40. Sun X, Han Y, Zhou L, Chen E, Lu B, Liu Y, Pan X, Cowley AW, Jr., Liang M,
Wu Q et al. A comprehensive evaluation of alignment software for reduced
representation bisulfite sequencing data. Bioinformatics 2018, 34(16):2715–​2723.
41. Zhou Q, Lim JQ, Sung WK, Li G. An integrated package for bisulfite DNA
methylation data analysis with indel-​sensitive mapping. BMC Bioinformatics
2019, 20(1):47.
42. Bock C. Analysing and interpreting DNA methylation data. Nat Rev Genet
2012, 13(10):705–​719.
43. Lin X, Sun D, Rodriguez B, Zhao Q, Sun H, Zhang Y, Li W. BSeQC: quality con-
trol of bisulfite sequencing experiments. Bioinformatics 2013, 29(24):3227–​3229.
44. MethylDackel (https://​git​hub.com/​dprya​n79/​Methy​lDac​kel)
45. Akalin A, Kormaksson M, Li S, Garrett-​Bakelman FE, Figueroa ME, Melnick
A, Mason CE. methylKit: a comprehensive R package for the analysis of
genome-​wide DNA methylation profiles. Genome Biol 2012, 13(10):R87.
46. Qu J, Zhou M, Song Q, Hong EE, Smith AD. MLML: consistent simultaneous
estimates of DNA methylation and hydroxymethylation. Bioinformatics 2013,
29(20):2645–​2646.
47. MethPipe (https://​git​hub.com/​smith​labc​ode/​methp​ipe/​)
48. Smith ZD, Chan MM, Mikkelsen TS, Gu H, Gnirke A, Regev A, Meissner
A. A unique regulatory phase of DNA methylation in the early mammalian
embryo. Nature 2012, 484(7394):339–​344.
49. Liu Y, Siegmund KD, Laird PW, Berman BP. Bis-​SNP: Combined DNA methy-
lation and SNP calling for Bisulfite-​seq data. Genome Biol 2012, 13(7):R61.
50. Washington University EpiGenome Browser (http://​epige​nome​gate​way.
wustl.edu/​brow​ser/​)
51. Kent WJ, Zweig AS, Barber G, Hinrichs AS, Karolchik D. BigWig and
BigBed: enabling browsing of large distributed datasets. Bioinformatics 2010,
26(17):2204–​2207.
52. Dorff KC, Chambwe N, Zeno Z, Simi M, Shaknovich R, Campagne F.
GobyWeb: simplified management and analysis of gene expression and DNA
methylation sequencing data. PLoS One 2013, 8(7):e69666.
53. Muller F, Scherer M, Assenov Y, Lutsik P, Walter J, Lengauer T, Bock C.
RnBeads 2.0: comprehensive analysis of DNA methylation data. Genome Biol
2019, 20(1):55.
54. Assenov Y, Muller F, Lutsik P, Walter J, Lengauer T, Bock C. Comprehensive
analysis of DNA methylation data with RnBeads. Nat Methods 2014,
11(11):1138–​1140.
55. Jiang P, Sun K, Lun FM, Guo AM, Wang H, Chan KC, Chiu RW, Lo YM, Sun H.
Methy-​Pipe: an integrated bioinformatics pipeline for whole genome bisulfite
sequencing data analysis. PLoS One 2014, 9(6):e100360.
56. Sun D, Xi Y, Rodriguez B, Park HJ, Tong P, Meong M, Goodell MA, Li W.
MOABS: model based analysis of bisulfite sequencing data. Genome Biol 2014,
15(2):R38.
57. Zhang Y, Liu H, Lv J, Xiao X, Zhu J, Liu X, Su J, Li X, Wu Q, Wang F et al.
QDMR: a quantitative method for identification of differentially methylated
regions by entropy. Nucleic Acids Res 2011, 39(9):e58.
Epigenomics by DNA Methylation Sequencing 339

58. Piao Y, Xu W, Park KH, Ryu KH, Xiang R. Comprehensive evaluation of dif-
ferential methylation analysis methods for bisulfite sequencing data. Int J
Environ Res Public Health 2021, 18(15):7975.
59. Liu Y, Han Y, Zhou L, Pan X, Sun X, Liu Y, Liang M, Qin J, Lu Y, Liu P. A com-
prehensive evaluation of computational tools to identify differential methyla-
tion regions using RRBS data. Genomics 2020, 112(6):4567–​4576.
60. Statham AL, Strbenac D, Coolen MW, Stirzaker C, Clark SJ, Robinson MD.
Repitools: an R package for the analysis of enrichment-​based epigenomic
data. Bioinformatics 2010, 26(13):1662–​1663.
61. Park Y, Figueroa ME, Rozek LS, Sartor MA. MethylSig: a whole genome DNA
methylation analysis pipeline. Bioinformatics 2014, 30(17):2414–​2422.
62. Dolzhenko E, Smith AD. Using beta-​binomial regression for high-​precision
differential methylation analysis in multifactor whole-​ genome bisulfite
sequencing experiments. BMC Bioinformatics 2014, 15:215.
63. Gaspar JM, Hart RP. DMRfinder: efficiently identifying differentially
methylated regions from MethylC-​seq data. BMC Bioinformatics 2017, 18(1):528.
64. Juhling F, Kretzmer H, Bernhart SH, Otto C, Stadler PF, Hoffmann S.
metilene: fast and sensitive calling of differentially methylated regions from
bisulfite sequencing data. Genome Res 2016, 26(2):256–​262.
65. Feng H, Conneely KN, Wu H. A Bayesian hierarchical model to detect differ-
entially methylated loci from single nucleotide resolution sequencing data.
Nucleic Acids Res 2014, 42(8):e69.
66. Halachev K, Bast H, Albrecht F, Lengauer T, Bock C. EpiExplorer: live explor-
ation and global analysis of large epigenomic datasets. Genome Biol 2012,
13(10):R96.
67. McLean CY, Bristor D, Hiller M, Clarke SL, Schaar BT, Lowe CB, Wenger
AM, Bejerano G. GREAT improves functional interpretation of cis-​regulatory
regions. Nat Biotechnol 2010, 28(5):495–​501.
15 
Whole Metagenome Sequencing for
Microbial Community Analysis

A small amount of environmental sample, such as a handful of soil, is rich


in microbial life, but the number of microbial species in such a sample is
unknown. The microbiome on or within our body contains tens of thousands,
if not more, species of bacteria, fungi, and archaea. Besides their tremendous
species diversity, the composition, as well as function, of such microbial
communities is not static but constantly changing according to the status of
their environment. Our current understanding of these diverse and dynamic
microbial communities is still significantly lacking, as most of our know-
ledge comes from culturable species. For those that still cannot be cultured in
the lab, which comprise the majority of microorganisms on earth, we know
very little. Metagenomics offers an important approach to study microbial
diversity in these environmental communities without relying on artifi-
cial culturing. Also referred to as environmental or community genomics,
metagenomics examines all genomes existing in a microbial community as a
whole without the need to capture or amplify individual genomes. Through
simultaneous analysis of all DNA molecules present in a microbial commu-
nity, metagenomics provides a profile of taxonomic composition and func-
tional status of the community and its environment.
Before the advent of NGS, metagenomics studies were usually conducted
with DNA cloning combined with Sanger sequencing. In this approach,
DNA extracted from a microbial community is first fragmented, and then the
DNA fragments are cloned into plasmid vectors for amplification in order
to produce enough materials for Sanger sequencing. With the continuous
development and significant cost drop in NGS technologies, massively par-
allel metagenomic sequencing has quickly replaced this traditional low-​
throughput approach and become a major approach for studying various
microbial communities. The high sensitivity offered by the NGS approach
provides direct access to the unculturable microbial majority that were pre-
viously “invisible” to analysis [1]. The Human Microbiome Project exempli-
fies the use of NGS in interrogating complex metagenomes, such as those at
different sites of the human body including the gastrointestinal tract. The
application of NGS in metagenomic analysis of a large variety of other micro-
bial communities, such as those in soil, the phyllosphere, the ocean, and those

DOI: 10.1201/9780429329180-18 341


342 Next-Generation Sequencing Data Analysis

associated with bioremediation and biofuel generation, has led to exponen-


tial increase in the number of metagenomes studied.
Compared to the NGS data generated from a single species (most chapters
in this book deal with individual species), the metagenomics data from micro-
bial community sequencing is much more complicated. Each metagenome
contains DNA sequences from a large but unknown number of species,
including viruses, bacteria, archaea, fungi, and microscopic eukaryotes. To
further complicate the situation, the relative abundance of these species
varies widely. In comparison to sequencing reads collected from a single
species, metagenomic sequencing reads contain much higher heterogeneity
because of the tremendous genome diversity in each microbial community.
Also because of the tremendous DNA sequence complexity contained in the
metagenome, most metagenomic sequencing effort can only sample part of
the DNA pool. As a result of this limited sampling in a highly diverse DNA
space, metagenomic NGS data is highly fragmented and has low redundancy.
Due to the lack of redundant (i.e., partially overlapping, not duplicate) reads,
metagenomic NGS data has an inherently higher error rate when compared
to single-​genome sequencing. All these differences between metagenomic
and monogenomic NGS data require an entirely different set of tools for
NGS-​based metagenome data analysis for microbial community compos-
itional and functional profiling.

15.1 Experimental Design and Sample Preparation


Metagenomics studies aim to determine identities and relative abundance
of different members, or taxa, in a microbial community, and how environ-
mental factors affect the composition and function of these communities. To
achieve this by sequencing, there are two general approaches: whole genome
shotgun metagenomic sequencing and targeted metagenomic sequencing.
The shotgun approach provides random sampling of all genomes contained
in an environmental or host-​ associated microbial sample. To carry out
shotgun sequencing, total DNA extracted from such a sample is first broken
into small fragments for short-​read sequencing, or high-​molecular-​weight
(HMW) DNA directly sequenced using long-​read sequencing.
In the targeted approach, genomic component(s) that are shared among
different species are PCR amplified and the amplicons are sequenced. The
most commonly used target in this approach is the 16S rRNA gene, while
other genes that code for specific protein functions (such as resistance to spe-
cific antibiotics) or non-​coding genes are also used. The 16S rRNA gene, being
considered as the universal clock of life [2], is usually used as a surrogate
marker for measuring the relative abundance of different operational taxo-
nomic units (OTUs, a metagenomics term to describe a species or a group of
Whole Metagenome Sequencing for Microbial Community Analysis 343

species when only DNA sequence information is available). By focusing on


the 16S rRNA gene or other specific genomic target(s), this approach greatly
reduces complexity in the generated data, thereby achieving deeper coverage
and accommodating more samples. It should be noted that the 16S rRNA-​
based approach only produces approximate estimation of relative taxonomic
abundance, due to 16S rRNA copy number variation in some species and the
fact that the standard 16S rRNA PCR primers may not bind to their supposed
target sites in all cases because of random mutation. In comparison, the
shotgun approach, while requiring significantly more sequencing, takes an
unbiased path to offer a comprehensive assessment of genome content in the
community, and thereby provides in-​depth information on community com-
position and function. This chapter focuses on shotgun metagenome sequen-
cing data analysis.

15.1.1 Metagenome Sample Collection


The success of a metagenomics project is to a degree dependent on factors
that are not related to genomics. One such factor is how much is known
about the habitat where study samples will be collected. The more physically,
chemically, and ecologically characterized the habitat is, the more knowledge
will be gathered from the metagenomic NGS data. In-​depth characteriza-
tion and detailed description of the sampling environment is one founda-
tion of a successful metagenomics experiment. Keeping detailed metadata
on the habitat and the sampling process, such as characteristics of the gen-
eral environment, geographical location and specific features of the sampling
locales, and the sampling method, is of great importance to downstream data
interpretation.
As the composition and complexity of a metagenome sample are
determined by the habitat and the sampling site, the unique characteristics of
a sampling environment, along with the question to be answered or specific
hypothesis to be tested, eventually determine how many reads are required.
It should also be emphasized that since where the samples are collected dir-
ectly shapes the outcome, the sampling sites must be representative of the
habitat under study. In order to collect representative samples, informa-
tion on spatial and temporal variation in the habitat must be known prior
to sample collection. If this information is not available, a small-​scale trial
shotgun sequencing run might prove helpful with a small number of samples
sequenced. Alternatively, a targeted 16S rRNA amplicon sequencing can also
be used to survey the diversity of the microbial community.

15.1.2 Metagenome Sample Processing


DNA extraction is the first and also a key step in metagenome sequencing
sample preparation. The DNA extracted from this step should represent
all, or at least most, members of the sampling community and their relative
344 Next-Generation Sequencing Data Analysis

abundance, be of high purity and free of contaminants that might interfere


with the subsequent sequencing library construction. While this step might be
routine in conventional genome sequencing for a single organism, extracting
high-​quality DNA from microbial community samples collected from various
habitats poses challenges. For example, humic acids, polysaccharides,
tannins, and other compounds are major contaminants in environmental
samples such as those from the soil, which if not removed can lead to inhib-
ition of enzymes used in library construction. In host-​associated habitats
such as the human gut, host DNA is the major potential contaminant.
Besides purity, extracting DNA in equal efficiency from different commu-
nity members is another challenge, as optimal condition of cell lysis for DNA
release from one group of microbes may not be ideal for another. For example,
mechanical disruption is often used for breaking up cells in metagenomics
studies, but by using this method DNA released from easily lysed cells may
be sheared to fragments when tougher cells are eventually disrupted. While
these challenging issues should be acknowledged and addressed, they are
not insurmountable and robust extraction protocols are available for various
habitats (e.g., [3]).
Advancements in sequencing library preparation protocols have reduced
the amounts of DNA required considerably to lower nanograms level (e.g.,
the Nextera XT protocol needs only 1 ng DNA to start). This should accom-
modate DNA extracted from most habitats. In situations where only very
limited amount of DNA is available, amplification of the DNA might be
needed to generate enough material for creating sequencing libraries. To
maintain the relative abundance level between community members, strat-
egies such as multiple displacement amplification can be used. Such ampli-
fication can generate more than enough DNA for library construction from
femtograms of starting DNA.

15.2 Sequencing Approaches
There are several key factors that need to be considered before the sequencing
process starts. These include sequencing depth, read length, and sequencing
platforms. The depth of sequencing is dependent on the species richness and
abundances in the samples, and the goal to be pursued. For example, a study
that attempts to locate rare members in a highly diverse microbial community
requires deeper sequencing than one that is only focused on more abundant
members in a less diverse environment. With regard to read length, longer
reads are always better than shorter reads in metagenomics for sorting out
the inherent sequence complexity. The read length from most current short
read sequencers can reach 150 bp from each end for paired-​end sequencing.
Whole Metagenome Sequencing for Microbial Community Analysis 345

As overviewed in Chapter 4, long-​read sequencing technologies, such as


PacBio’s SMRT and ONT’s nanopore sequencing, generate much longer
(but fewer and less accurate) reads. With read throughput and sequencing
accuracy steadily increasing, there are increasing numbers of samples being
sequenced on these systems. A hybrid approach is also often used to take
advantage of the different strength of these technologies, with the use of short
reads to generate an in-​depth survey of the community and long reads to
provide scaffolding for assembling contigs (see next). Future advancements
in sequencing technologies will undoubtedly lead to continuous increase
in read length and drop in cost making the goals of metagenomics more
achievable.

15.3 Overview of Shotgun Metagenome


Sequencing Data Analysis
For microbial community profiling, whole-​metagenome shotgun sequen-
cing provides rich information on a community’s taxonomic composition
and functional status, without requiring pre-​ existing knowledge of all
genomes contained in the community. Figure 15.1 shows an overview of
a general shotgun metagenomic sequencing data analysis workflow. Most
of this workflow involves assembly of reads to reconstruct the so-​called
metagenome-​ assembled genomes (or MAGs), while an assembly-​ free
approach uses sequencing reads directly. For both approaches, to perform
taxonomic profiling and functional analysis, sequence homology and other
feature search against genes of microbes in the currently known taxonomy,
as catalogued in various public databases, are key steps. While results from
these key steps are limited to the currently known genomes and catalogued
sequences, the rapid increase in the number of sequenced microbial genomes
will gradually alleviate this limitation. The rapid increase in the employ-
ment of long-​read sequencing to the metagenomics field also helps with
reconstruction of more MAGs for subsequent analyses. Besides taxonomic
profiling and functional analysis in one condition or habitat, comparative
metagenomics analysis between conditions or habitats is usually performed
to achieve the final goal of studying the effects of environmental factors on
a microbial community.
The following sections cover the various steps of metagenomics data ana-
lysis. Because of the great diversity in sampling habitats/​conditions and
the specific questions asked in each study, there is no fixed workflow for
metagenomics data analysis. The steps outlined in Figure 15.1 and covered
next are not necessarily arranged in the most appropriate order for a par-
ticular project, and they can be used in different combinations and/​or with
346 Next-Generation Sequencing Data Analysis

Metagenome Sample Collection


& Metadata Recording

Sample Processing
& Sequencing

Data QC & Preprocessing

Reads Binning

Assembly-Free Assembly-Dependent

Metagenome Assembly
Reads Mapping to Currently
Known Gene Sequences
Contig Binning

Calling of Genes/
Genomic Elements

Taxonomic Profiling Gene Function Annotation

Comparative Metagenomic Functional Profiling/Metabolic


Analysis Pathway Reconstruction

FIGURE 15.1
Major steps of metagenome analysis.

some step(s) omitted. Since the publication of the first edition of this book,
there has been rapid increase in the number of tools available for metagenome
analysis. Some of the currently available tools, such as those required for
taxonomic profiling based on search against multiple databases, require con-
siderable computing resources and power.
Whole Metagenome Sequencing for Microbial Community Analysis 347

15.4 Sequencing Data Quality Control and Preprocessing


To ensure data quality and avoid erroneous results, metagenomic shotgun
sequencing reads should first be examined and preprocessed prior to
conducting downstream analysis. Using the tools introduced in Chapter 5,
reads of low quality should be filtered out, and low-​quality bases and adapter
sequences trimmed off. In addition, for samples from host-​associated habitats,
contaminating host sequences need to be marked and excluded from further
analysis. This can be realized by simply using mappers such as bowtie2 (the -​-​
un-​conc output option), or using bowtie2 to output all reads and then samtools
to extract reads unmapped to the host genome. Specially developed tools for
marking and removing DNA contamination sequences include KneadData
[4], BMTagger [5], and DeconSeq [6]. Additional data preprocessing also
includes removal of duplicated reads. This can be conducted with tools such
as the Dedupe tool in the BBTools suite [7], FastUniq [8], and the Picard
module called EstimateLibraryComplexity, all of which identify and remove
duplicate reads without the need to align reads to a reference genome.

15.5 Taxonomic Characterization of a Microbial Community


15.5.1 Metagenome Assembly
While the ultimate goal of metagenomics is to assemble each genome in a
microbial community, this is currently still far from achievable because of
several reasons. The number of organisms in a metagenome is unknown, and
there are wide variations in their relative abundance and therefore sequen-
cing depth among the organisms. This is especially the case for samples
collected from highly complex microbial communities. The large number of
species in these samples, and the concomitant low sequencing depth for most
species, make metagenome assembly extremely challenging. Sequence simi-
larity between closely related species poses further challenges to assemblers,
often leading to chimeric assemblies that contain reads from different OTUs.
Despite the challenges, metagenome reads assembly is an important step in
metagenomic sequencing data analysis. It has led to rapid increase in the
total number of MAGs recovered from a wide range of environments, such
as the human gut [9, 10], and a diversity of habitats on Earth encompassing
all continents and oceans [11].
For de novo metagenome assembly, the assemblers introduced in Chapter 12
for single-​genome de novo assembly, such as SOAPdenovo and Velvet, were
initially applied but with limited success. As a result, assemblers tailored for
metagenome reads have been developed. For assembling long reads such
348 Next-Generation Sequencing Data Analysis

as those generated from the PacBio and ONT platforms, assemblers such as
metaFlye [12], Raven [13], Canu [14], and Hifiasm-​meta [15] can be used. For
short reads, more assemblers are available, including SPAdes/​metaSPAdes
[16, 17], MEGAHIT [18], IDBA-​UD [19], MetaVelvet/​MetaVelvet-​SL [20, 21],
and Ray Meta [22]. Similar to single-​genome assemblers, many of these short
read metagenome assemblers, such as metaSPAdes, MEGAHIT, IDBA-​UD,
and Ray Meta, are based on the de Bruijn graph approach (see Chapter 12).
In addition, these methods use multi-​k-​mer sizes, instead of a fixed k-​mer
size, in order to improve assemblies. The difference from the single-​genome
assemblers, though, is that they attempt to identify subgraphs within a
mixed de Bruijn graph, each of which is expected to represent an individual
genome. For example, metaSPAdes first builds a large de Bruijn graph from
all metagenomic reads using SPAdes and then transforms it into an assembly
graph. Within the assembly graph, subgraphs that contain alternative paths
are identified, corresponding to large fragments from individual genomes.
Besides these assemblers that are designed for either long or short reads, other
assemblers combine long and short reads in an effort to increased assembly
quality. These hybrid assemblers include MaSuRCA [23], hybridSPAdes [24],
and OPERA-​MS [25].
After the assembly process, a metagenome usually comprises mostly of
contigs of various sizes. To evaluate the assembly quality, traditional evalu-
ation metrics, such as N50, are not as informative and representative as in
evaluating single-​genome assemblies. Instead, aggregate statistics such as
the total number of contigs, the percentage of reads mapped to them, and
the maximum, median, and average lengths of the contigs are often used.
Further inspection of the assembly quality includes looking for chimeric or
mis-​assemblies. There are currently a number of tools available to assess MAG
quality, including CheckM [26], MetaQUAST [27], and BUSCO [28], all of
which rely on reference genomes. Reference-​free tools include DeepMAsED
[29] and ALE (Assembly Likelihood Evaluation) [30].
After contig assembly, if paired reads are available, metagenome scaffolds
can be built from the contigs. Many of the metagenome assemblers have
a module to carry out scaffolding. Besides these modules, dedicated
metagenome scaffolding tools like Bambus 2 [31] may be used to determine if
additional scaffolding is needed. Bambus 2 accepts contigs constructed with
most assemblers using reads from all sequencing platforms. In the process
of building scaffolds from contigs, ambiguous and inconsistent contigs may
also be identified. Besides scaffolding, another approach for the assembly of
MAGs is contig binning, which places contigs derived from the same genome
into the same bin. Contigs in the same bin are then reassembled into a MAG.

15.5.2 Sequence Binning
As indicated above, metagenomic sequence binning refers to the pro-
cess of clustering sequence fragments in a mixture into different “bins”
Whole Metagenome Sequencing for Microbial Community Analysis 349

corresponding to their genomic or taxonomic origins. This process can be


conducted on contigs (most often), reads, or genes. With longer reads or
contigs, high-​resolution binning can be achieved at the levels of family,
genus, species, or even strains. Short sequences may be binned only to the
level of phylum due to the limited information carried in the sequences. Since
sequence binning reduces the complexity inherent in the metagenomics data,
each set of binned sequences can then be subjected to independent analysis
in other steps, such as MAG reconstruction for identification of new archae-
bacterial and eubacterial specie or viruses.
Two binning approaches are usually used. One approach, called genome
binning, bins contigs or reads on the basis of sequence composition and
abundance. Sequence composition encompasses characteristics such as oligo-
nucleotide (often tetramer) frequency, G/​C content, and codon usage. The
genome binning approach is built on the assumption that sequences from the
same species or closely related species/​organisms are more similar to each
other in these characteristics and abundance than to distantly or non-​related
species/​organisms. Methods that use this strategy include MetaBAT 2 [32],
MaxBin 2.0 [33], GroopM [34] (now replaced by Rosella) [35], CONCOCT
[36], MetaWatt [37], and VAMB [38] (Table 15.1). CONCOCT, as an example
for genome binning, places contigs into bins based on sequence composition
and coverage across samples. After combining k-​mer frequency and coverage
information, CONCOCT applies PCA to reduce the dimensionality of the
matrix and reveal clusters. This is followed by the utilization of a Gaussian
mixture model (GMM) for binning the contigs. VAMB is another example
of genome binning method that is based on machine learning. It uses deep
variational autoencoders to encode sequence co-​abundance and k-​mer distri-
bution before clustering of sequences. Besides the individual methods, there
are also ensemble methods that integrate binning results from the different
individual genome binning methods. Examples of such ensemble binning
methods are MetaBinner [39], MetaWRAP [40], and DAS tool [41]. The
Critical Assessment of Metagenome Interpretation (CAMI) challenge round
II shows that some of these ensemble methods can achieve better results than
individual methods [42]. In general, the genome binning approach is more
reliable for contigs and long reads, as short reads carry less information due
to their limited length. While it has the advantage of being fast as it does
not rely on aligning metagenomic sequences to reference databases, vari-
ation in the distribution of sequence composition and coverage can lead to
inaccuracies.
The other approach, called taxonomy binning, assigns metagenomic
sequences to their taxonomic sources of origin by comparing against micro-
bial sequence database(s) that are taxonomically annotated. Methods that use
this approach first place sequences into different bins, followed by labeling of
the bins with taxonomic identifiers. Examples of taxonomy binning methods
include MEGAN/​MEGAN-​LR (for long reads) [43, 44], Kraken 2 [45], and
PhyloPythiaS+​ [46] (Table 15.1). MEGAN (for Metagenome Analyzer), as
350 Next-Generation Sequencing Data Analysis

an example, first aligns reads against a reference database such as BLAST-​


nr using an alignment tool called DIAMOND [47] that achieves fast
alignment speed. The alignment result is then processed by a program called
MEGANIZER for taxonomic binning that aims to assign a taxon ID to each
sequence. This binning process uses an algorithm called naïve LCA, which
assigns each read to a node representing the lowest common ancestor. Based
on this algorithm, a read that aligns to a widely conserved gene is assigned

TABLE 15.1
Commonly Used Binning Algorithms
Name Description Reference

Genome Binning
MaxBin 2.0 Classifies contigs into different bins using an Expectation-​ [33]
Maximization algorithm on the basis of their tetranucleotide
frequencies and coverages
CONCOCT Combines coverage and tetramer frequency for contig binning [36]
using GMM
MetaBAT 2 Uses adaptive binning to group the most reliable contigs [32]
first (such as those of high length), and then gradually add
remaining contigs
MetaWatt Uses multivariate statistics of tetramer frequencies and [37]
differential coverage information for binning. Also assesses
binning quality using taxonomic annotation of contigs in
each bin
VAMB A machine-​learning based binner that encodes k-​mer [38]
distribution and sequence co-​abundance information using
variational autoencoders for subsequent binning
GroopM Bins sequences by primarily leveraging differential coverage [34]
information
MetaBinner An ensemble binner that integrates component binning results [39]
generated with multiple features and initiations
MetaWRAP An ensemble binner to generate hybrid bin sets from other [40]
binners and select final bins based on CheckM results
Taxonomy Binning
MEGAN Aligns sequences against the NCBI-​nr reference database [43]
and then performs taxonomic binning using the naïve LCA
algorithm
Kraken 2 Assigns taxonomic labels to sequences based on search of [45, 48]
k-​mers within the sequences against a database of indexed
and sorted k-​mers (or their minimizers) extracted from all
genomes
PhyloPythiaS+​ Achieves taxonomic binning through building sample-​ [46]
specific support vector machine taxonomic classifier using
most relevant taxa and training sequences determined
automatically
Whole Metagenome Sequencing for Microbial Community Analysis 351

to a taxon at a higher level (e.g., phylum or class), while another read that
aligns to a less conserved gene that is limited to a select group of organisms
is assigned to a lower-​level taxon (such as genus or species). As it is based on
the current annotation of catalogued sequences, this approach is not suitable
to find currently unknown species or taxa.

15.5.3 Calling of Genes and Other Genomic Elements


from Metagenomic Sequences
To answer the questions of what taxonomic groups are in a microbial commu-
nity and what they are doing, identification of genes from assembled contigs
or MAGs is an essential step. For gene coding region identification, since
ORF-​containing metagenomic sequences may not carry full-​length ORFs,
metagenome ORF-​calling algorithms do not penalize for their incomplete-
ness. Many metagenome ORF callers employ machine learning strategies. For
example, Prodigal [49] and MetaGeneAnnotator [50] employ dynamic pro-
gramming. GeneMarkS-​2 [51], FragGeneScan (FGS) [52], and Glimmer-​MG
[53] are based on Markov models. CNN-​MGP [54] and Balrog [55] use con-
volutional neural networks. Among these methods, Prodigal is an unsuper-
vised algorithm that does not require training data. It predicts protein-​coding
genes using inherent properties in the sequences themselves, such as start
codon usage, ribosomal-binding site motif usage, genetic code usage, G/​C
content, and hexamer coding statistics, among other information. Calling of
other genomic elements, such as ncRNAs and CRISPRs, may require long
reads or contigs as well as more computational resources. Currently a limited
number of tools are available to identify these elements, such as tRNAscan-​
SE [56], ARAGORN [57], MinCED (a modified version of the CRISPR
Recognition Tool or CRT) [58], CRISPRFinder [59], and CRISPRDetect [60].
Besides providing answers to the composition and function of a microbial
community, calling of genes and other genomic elements also helps identify
mis-​assembled reads or locate adjoining contigs that are not yet placed into
the same scaffold.

15.5.4 Taxonomic Profiling
One goal of metagenome analysis is profile taxonomic composition and rela-
tive abundance of each taxon in a microbial community. This is related to
but different from taxonomic binning, which aims to group metagenomic
sequences into different bins. The CAMI II challenge finds that taxonomic
profilers MetaPhlAn [61] and mOTUs [62] had the best overall perform-
ance, both of which are based on the use of phylogenetic gene markers not
taxonomic binning of reads. Phylogenetic gene markers are composed of
ubiquitous but phylogenetically diverse genes, with good examples being
the rRNA genes (e.g., 16S), recA (DNA recombinase A), rpoB (RNA poly-
merase beta subunit), fusA (protein chain elongation factor), and gyrB (DNA
352 Next-Generation Sequencing Data Analysis

gyrase subunit B). MetaPhlAn, as an example, conducts homology search of


metagenomic reads against an extensive list of clade-​specific gene markers.
From the mapping of query reads to the marker genes, the number of
mapped reads is normalized prior to calculation of relative abundance levels
of different taxa identified in the sample [61]. Other commonly used taxo-
nomic profilers include Kaiju [63], Centrifuge [64], Bracken [65], GTDB-​tk
[66], and IGGsearch [9]. Among these tools, GTDB-​tk is a toolkit that assigns
taxonomic classification of MAGs using the Genome Taxonomy Database
(GTDB). It first calls genes from MAGs using Prodigal, then identifies marker
genes, followed by subsequent marker-​gene-​based phylogenetic inference
[67]. Bracken, which is built upon taxonomic binning results from Kraken,
uses a Bayesian approach to estimate the abundance levels of different taxa.
Taxonomic profilers developed specifically for long reads include MetaMaps
[68] and BugSeq [69]. Currently available comparisons on the use of such
long-​read taxonomic profilers vs. the aforementioned short-​read methods
suggest that the long-​range sequence information provided by long reads
and the methods designed to take advantage of such information lead to
improved taxonomic profiling [70].

15.6 Functional Characterization of a Microbial Community


15.6.1 Gene Function Annotation
Gene calling from metagenomic sequences provides substrate for functional
analysis of the underlying community, i.e., answering the question of “what
they are doing.” Functional annotation of called genes can reveal the full rep-
ertoire of protein functions in a habitat, including metabolism, signal trans-
duction, stress tolerance, virulence, etc. Uncommon functions may suggest
unusual lifestyle and activity in a community. The relative abundance of
different types of genes also reveals specificity about a community and how
organisms in the community deal with environmental factors in the habitat.
To conduct functional annotation, predicted protein sequences from called
ORFs are searched against a database of reference protein sequences, or
HMMs that describe protein families. Protein sequence databases such as
UniProt [71], InterPro [72], COG [73], and eggNOG [74], are among the most
commonly used databases. This task of database searching to identify all pos-
sible peptides coded by the metagenome is a computationally intensive pro-
cess. If local computing resources permit, locally installed stand-alone tools
such as Prokka [75], DRAM [76], DFAST [77], or NCBI’s Prokaryotic Genome
Annotation Pipeline (PGAP) [78] can be used. Alternatively, the task can
also be submitted to a web-​based system such as the metagenomics RAST
server (MG-​RAST) [79], the Integrated Microbial Genomes & Microbiomes
Whole Metagenome Sequencing for Microbial Community Analysis 353

system (IMG/​M) [80], GhostKOALA [81], or MGnify [82]. Some tools, such
as eggNOG-​mapper [83] provide both online and stand-alone versions.

15.6.2 Gene Function Profiling and Metabolic Pathway Reconstruction


To further answer the question of “what they are doing,” functional profiling
of a microbial community can be performed at the levels of annotated genes
(or gene families) and metabolic pathways. Analysis at the gene/​gene family
level relies on function annotations with the use of the databases detailed
above, while at the metabolic pathway level this requires databases that cap-
ture currently known metabolic pathways, including KEGG [84] and MetaCyc
[85]. Based on reference information from these databases, functional pro-
filing tools such as HUMAnN [61] and Carnelian [86] provide the abundance
of each gene/​gene family, or metabolic pathway detected in a community.
This abundance profile can be further stratified to reveal the contribution of
each identified OTU in the community. As an example, HUMAnN employs
MBLASTX to search metagenomic reads against the KEGG Orthology data-
base to determine the abundances of individual orthologous protein families.
At the pathway level, HUMAnN reconstructs pathways using MinPath [87],
which is a maximum parsimony approach to explain the observed families
and their abundances with a minimal set of pathways. After further noise
reduction and smoothing, the output from HUMAnN displays pathway
coverage (i.e., whether each pathway is present or absent), and the relative
abundance of each pathway in the metagenomic samples.
Besides quantifying present metabolic pathways, continuous methods
development also makes it possible to semi-​ automatically reconstruct
genome-​scale metabolic models, or GSMMs, to reveal the metabolic poten-
tial of a genome. This reconstruction process requires integration of gene
coding information in an assembled individual microbial genome and meta-
bolic pathway information catalogued in reference databases. Examples of
such methods are ModelSEED [88], CarveMe [89], Pathway Tools [90], Merlin
[91], and RAVEN [92]. From the reconstructed single-​species GSMMs and the
relative species abundance measured from a metagenomic sample, the many
GSMMs can be integrated to predict the metabolic status of the underlying
community. MAMBO, for Metabolomic Analysis of Metagenomes using fBa
and Optimization, is a good exemplary tool for this task [93].
In addition, the increasing availability of metabolomics data also makes
it possible to correlate metagenomic sequence and metabolite data. For
example, MIMOSA is a reference-​based tool to correlate metagenomic and
metabolomic data [94]. With this tool, community metabolic potential scores
can be calculated from metagenomic sequencing data to describe the potential
ability of a microbial community to metabolize small molecules. Abundances
of metabolites, as measured by metabolomics, can then be correlated with
the community’s metabolic potential scores. Furthermore, the specific taxa,
354 Next-Generation Sequencing Data Analysis

genes, or reactions that may contribute to the formation of the metabolites


can be identified. MelonnPan is another tool designed to predict metabolomic
profiles from microbial sequencing features. This method does not rely on
reference database search, instead it uses a machine learning approach [95].

15.7 Comparative Metagenomic Analysis


Comparative metagenomic analysis between habitats or conditions can lead
to insights about the underlying microbial communities and their dynamics.
However, statistical comparison between metagenomes is not as straightfor-
ward as other NGS-​based comparative analyses (such as RNA-​seq). This is
mostly due to the tremendous amount of variability involved in comparative
metagenomic analysis. One source of this variability is biological, as microbial
composition can vary greatly between different samples. Another source is
technical, due to insufficient sequencing depth and therefore undersampling
of low-​abundance species. These species generate fewer reads and are more
affected by stochastic factors in the sequence sampling process, as in general the
number of reads from a species is dependent on a number of factors, including
relative abundance of the species, genome size, genome copy number, within-​
species heterogeneity, and DNA extraction efficiency. Due to these biological
and technical factors, many species or OTUs detected in one sample or con-
dition are often absent in another sample or condition. If rare species need
to be studied in a metagenome study, it is more cost effective to artificially
increase their abundance using cell enrichment technologies such as flow cell
sorting rather than increasing sequencing depth. In a typical metagenomics
project that does not artificially increase the abundance of rare species, their
undersampling can lead to significant biases in subsequent data normaliza-
tion and detection of significant differences between samples. Compared to
other steps in the metagenomic data analysis pipeline, there has been rela-
tively less method development in comparative metagenomic analysis.

15.7.1 Metagenome Sequencing Data Normalization


Similar to RNA-​ seq data, metagenomic abundance data needs to be
normalized prior to comparative analysis. Currently there is still no consensus
as to how metagenomics data should be normalized. Among the normaliza-
tion approaches that have been reported, total-​sum scaling (TSS), equivalent
to the Total Count approach in RNA-​seq (Chapter 7), is performed by div-
iding the raw count of reads assigned to a certain species or OTU by the total
number of reads in the same sample. Another approach is cumulative-​sum
scaling (CSS), which, similar to the Upper Quartile approach in RNA-​seq, is
calculated by dividing the raw count of reads assigned to a species or OTU by
Whole Metagenome Sequencing for Microbial Community Analysis 355

the cumulative sum of counts up to a certain percentile. Other normalization


methods introduced in Chapter 7, such as TMM and RLE, can also be used on
metagenomic abundance data.

15.7.2 Identification of Differentially Abundant Species or OTUs


To identify species or OTUs that are differentially abundant between habitats
or conditions, currently available tools include metagenomeSeq [96], phyloseq
[97], LEfSe [98], STAMP [99], ANCOM and ANCOM-​BC [100, 101], corncob
[102], and MaAsLin 2 [103]. These tools use different methods and statistics
to detect differential abundance between metagenomes. For example,
metagenomeSeq implements the CSS normalization, and a distribution mix-
ture statistical model to deal with the biases caused by the undersampling
issue that confound comparative metagenomic analyses. LEfSe uses the
Kruskal–​Wallis rank-​sum test to detect features that display significant differ-
ential abundance between conditions. Among the newer methods, MaAsLin
2 uses general linear models to detect associations between abundances
of microbial features, such as taxa (or genes), and environmental or other
phenotypic metadata. It offers a number of normalization methods including
TSS, CSS, TMM, etc.

15.8 Integrated Metagenomics Data Analysis Pipelines


Besides the tools developed for each of the individual steps above, pipelines
designed for integrated comprehensive analysis of metagenomics data are
also available. These pipelines, including Quantitative Insight Into Microbial
Ecology (QIIME2) [104], IMG/​ M, MEGAN, MG-​ RAST, metaWRAP, and
bioBakery [61], contain large collections of tools that encompass the many
aspects of metagenomics data mining from preprocessing, binning, feature
identification, functional annotation, to cross-​condition comparison. For
example, MG-​RAST directly takes sequencing and metadata files as input,
conducts reads QC and preprocessing, gene calling, protein identification,
annotation mapping, abundance profiling, comparative analysis, and meta-
bolic reconstruction.

15.9 Metagenomics Data Repositories


In the United States, like for other NGS data, the NCBI SRA database provides
the official repository for all metagenomic data collected by NGS technologies.
356 Next-Generation Sequencing Data Analysis

In Europe, MGnify (previously EBI Metagenomics) offers archiving and ana-


lysis of metagenomics data. The data archived by MGnify is also accessible
through ENA (or SRA). In China, a database called gcMeta, or Global Catalogue
of Metagenomics [105], has been established more recently. Besides these offi-
cial metagenomics data repositories, MG-​RAST and IMG/​M are two de facto
metagenomic data repositories that also enable data sharing in a collabora-
tive environment and with the entire research community. The value of these
repositories will become more apparent when more and more metagenomics
data becomes available. For example, they can accelerate the discovery of
new genes and species through providing opportunities to compare currently
unknown sequences that exist in multiple metagenomes. In a typical shotgun
metagenomics study, many sequences are previously unknown and may
represent novel genes or sequences from currently uncatalogued species. To
discover novel genes and new species, meta-​analysis of data (including meta-
data) is needed, which is only enabled by these repositories.

References
1. Lloyd KG, Steen AD, Ladau J, Yin J, Crosby L. Phylogenetically novel
uncultured microbial cells dominate earth microbiomes. mSystems 2018,
3(5):e00055-​18.
2. Yarza P, Ludwig W, Euzeby J, Amann R, Schleifer KH, Glockner FO, Rossello-​
Mora R. Update of the All-​Species Living Tree Project based on 16S and 23S
rRNA sequence analyses. Syst Appl Microbiol 2010, 33(6):291–​299.
3. Delmont TO, Robe P, Clark I, Simonet P, Vogel TM. Metagenomic comparison
of direct and indirect soil DNA extraction approaches. J Microbiol Methods
2011, 86(3):397–​400.
4. McIver LJ, Abu-​Ali G, Franzosa EA, Schwager R, Morgan XC, Waldron L,
Segata N, Huttenhower C. bioBakery: a meta’omic analysis environment.
Bioinformatics 2018, 34(7):1235–​1237.
5. BMTagger (http://​biow​ulf.nih.gov/​apps/​bmtag​ger.html)
6. Schmieder R, Edwards R. Fast identification and removal of sequence contam-
ination from genomic and metagenomic datasets. PLoS One 2011, 6(3):e17288.
7. Bushnell B. BBMap: a fast, accurate, splice-​ aware aligner. In.: Lawrence
Berkeley National Lab.(LBNL), Berkeley, CA (United States); 2014.
8. Xu H, Luo X, Qian J, Pang X, Song J, Qian G, Chen J, Chen S. FastUniq: a
fast de novo duplicates removal tool for paired short reads. PLoS One 2012,
7(12):e52249.
9. Nayfach S, Shi ZJ, Seshadri R, Pollard KS, Kyrpides NC. New insights from
uncultivated genomes of the global human gut microbiome. Nature 2019,
568(7753):505–​510.
10. Almeida A, Mitchell AL, Boland M, Forster SC, Gloor GB, Tarkowska A,
Lawley TD, Finn RD. A new genomic blueprint of the human gut microbiota.
Nature 2019, 568(7753):499–​504.
Whole Metagenome Sequencing for Microbial Community Analysis 357

11. Nayfach S, Roux S, Seshadri R, Udwary D, Varghese N, Schulz F, Wu D,


Paez-​Espino D, Chen IM, Huntemann M et al. A genomic catalog of Earth’s
microbiomes. Nat Biotechnol 2021, 39(4):499–​509.
12. Kolmogorov M, Bickhart DM, Behsaz B, Gurevich A, Rayko M, Shin SB,
Kuhn K, Yuan J, Polevikov E, Smith TPL et al. metaFlye: scalable long-​
read metagenome assembly using repeat graphs. Nat Methods 2020, 17(11):
1103–​1110.
13. Vaser R, Šikić M. Time-​and memory-​efficient genome assembly with Raven.
Nat Comput Sci 2021, 1(5):332–​336.
14. Koren S, Walenz BP, Berlin K, Miller JR, Bergman NH, Phillippy AM.
Canu: scalable and accurate long-​read assembly via adaptive k-​mer weighting
and repeat separation. Genome Res 2017, 27(5):722–​736.
15. Feng X, Cheng H, Portik D, Li H. Metagenome assembly of high-​fidelity long
reads with Hifiasm-​meta. Nat Methods 2022, 19(6):671–​674.
16. Nurk S, Meleshko D, Korobeynikov A, Pevzner PA. metaSPAdes: a new versa-
tile metagenomic assembler. Genome Res 2017, 27(5):824–​834.
17. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS,
Lesin VM, Nikolenko SI, Pham S, Prjibelski AD et al. SPAdes: a new genome
assembly algorithm and its applications to single-​cell sequencing. J Comput
Biol 2012, 19(5):455–​477.
18. Li D, Liu CM, Luo R, Sadakane K, Lam TW. MEGAHIT: an ultra-​fast single-​
node solution for large and complex metagenomics assembly via succinct de
Bruijn graph. Bioinformatics 2015, 31(10):1674–​1676.
19. Peng Y, Leung HC, Yiu SM, Chin FY. IDBA-​UD: a de novo assembler for
single-​
cell and metagenomic sequencing data with highly uneven depth.
Bioinformatics 2012, 28(11):1420–​1428.
20. Namiki T, Hachiya T, Tanaka H, Sakakibara Y. MetaVelvet: an extension of
Velvet assembler to de novo metagenome assembly from short sequence
reads. Nucleic Acids Res 2012, 40(20):e155.
21. Afiahayati, Sato K, Sakakibara Y. MetaVelvet-​SL: an extension of the Velvet
assembler to a de novo metagenomic assembler utilizing supervised learning.
DNA Res 2015, 22(1):69–​77.
22. Boisvert S, Raymond F, Godzaridis E, Laviolette F, Corbeil J. Ray
Meta: scalable de novo metagenome assembly and profiling. Genome Biol
2012, 13(12):R122.
23. Zimin AV, Marcais G, Puiu D, Roberts M, Salzberg SL, Yorke JA. The MaSuRCA
genome assembler. Bioinformatics 2013, 29(21):2669–​2677.
24. Antipov D, Korobeynikov A, McLean JS, Pevzner PA. hybridSPAdes: an
algorithm for hybrid assembly of short and long reads. Bioinformatics 2016,
32(7):1009–​1015.
25. Bertrand D, Shaw J, Kalathiyappan M, Ng AHQ, Kumar MS, Li C, Dvornicic
M, Soldo JP, Koh JY, Tong C et al. Hybrid metagenomic assembly enables high-​
resolution analysis of resistance determinants and mobile elements in human
microbiomes. Nat Biotechnol 2019, 37(8):937–​944.
26. Parks DH, Imelfort M, Skennerton CT, Hugenholtz P, Tyson GW.
CheckM: assessing the quality of microbial genomes recovered from isolates,
single cells, and metagenomes. Genome Res 2015, 25(7):1043–​1055.
27. Mikheenko A, Saveliev V, Gurevich A. MetaQUAST: evaluation of metagenome
assemblies. Bioinformatics 2016, 32(7):1088–​1090.
358 Next-Generation Sequencing Data Analysis

28. Simao FA, Waterhouse RM, Ioannidis P, Kriventseva EV, Zdobnov EM.
BUSCO: assessing genome assembly and annotation completeness with
single-​copy orthologs. Bioinformatics 2015, 31(19):3210–​3212.
29. Mineeva O, Rojas-​ Carulla M, Ley RE, Scholkopf B, Youngblut ND.
DeepMAsED: evaluating the quality of metagenomic assemblies. Bioinformatics
2020, 36(10):3011–​3017.
30. Clark SC, Egan R, Frazier PI, Wang Z. ALE: a generic assembly likelihood
evaluation framework for assessing the accuracy of genome and metagenome
assemblies. Bioinformatics 2013, 29(4):435–​443.
31. Koren S, Treangen TJ, Pop M. Bambus 2: scaffolding metagenomes.
Bioinformatics 2011, 27(21):2964–​2971.
32. Kang DD, Li F, Kirton E, Thomas A, Egan R, An H, Wang Z. MetaBAT 2: an
adaptive binning algorithm for robust and efficient genome reconstruction
from metagenome assemblies. PeerJ 2019, 7:e7359.
33. Wu YW, Simmons BA, Singer SW. MaxBin 2.0: an automated binning algo-
rithm to recover genomes from multiple metagenomic datasets. Bioinformatics
2016, 32(4):605–​607.
34. Imelfort M, Parks D, Woodcroft BJ, Dennis P, Hugenholtz P, Tyson GW.
GroopM: an automated tool for the recovery of population genomes from
related metagenomes. PeerJ 2014, 2:e603.
35. Rosella (https://​git​hub.com/​rhy​snew​ell/​rose​lla)
36. Alneberg J, Bjarnason BS, de Bruijn I, Schirmer M, Quick J, Ijaz UZ, Lahti
L, Loman NJ, Andersson AF, Quince C. Binning metagenomic contigs by
coverage and composition. Nat Methods 2014, 11(11):1144–​1146.
37. Strous M, Kraft B, Bisdorf R, Tegetmeyer HE. The binning of metagenomic
contigs for microbial physiology of mixed cultures. Front Microbiol 2012, 3:410.
38. Nissen JN, Johansen J, Allesoe RL, Sonderby CK, Armenteros JJA, Gronbech
CH, Jensen LJ, Nielsen HB, Petersen TN, Winther O et al. Improved
metagenome binning and assembly using deep variational autoencoders. Nat
Biotechnol 2021, 39(5):555–​560.
39. Wang Z, Huang P, You R, Sun F, Zhu S. MetaBinner: a high-​performance and
stand-​alone ensemble binning method to recover individual genomes from
complex microbial communities. Genome Biol 2023, 24(1):1.
40. Uritskiy GV, DiRuggiero J, Taylor J. MetaWRAP-​a flexible pipeline for genome-​
resolved metagenomic data analysis. Microbiome 2018, 6(1):158.
41. Sieber CMK, Probst AJ, Sharrar A, Thomas BC, Hess M, Tringe SG, Banfield JF.
Recovery of genomes from metagenomes via a dereplication, aggregation and
scoring strategy. Nat Microbiol 2018, 3(7):836–​843.
42. Meyer F, Fritz A, Deng ZL, Koslicki D, Lesker TR, Gurevich A, Robertson
G, Alser M, Antipov D, Beghini F et al. Critical Assessment of Metagenome
Interpretation: the second round of challenges. Nat Methods 2022, 19(4):429–​440.
43. Huson DH, Beier S, Flade I, Gorska A, El-​Hadidi M, Mitra S, Ruscheweyh
HJ, Tappu R. MEGAN Community Edition –​Interactive Exploration and
Analysis of Large-​Scale Microbiome Sequencing Data. PLoS Comput Biol 2016,
12(6):e1004957.
44. Huson DH, Albrecht B, Bagci C, Bessarab I, Gorska A, Jolic D, Williams RBH.
MEGAN-​LR: new algorithms allow accurate binning and easy interactive
exploration of metagenomic long reads and contigs. Biol Direct 2018, 13(1):6.
Whole Metagenome Sequencing for Microbial Community Analysis 359

45. Wood DE, Lu J, Langmead B. Improved metagenomic analysis with Kraken 2.


Genome Biol 2019, 20(1):257.
46. Gregor I, Droge J, Schirmer M, Quince C, McHardy AC. PhyloPythiaS+​: a self-​
training method for the rapid reconstruction of low-​ranking taxonomic bins
from metagenomes. PeerJ 2016, 4:e1603.
47. Buchfink B, Xie C, Huson DH. Fast and sensitive protein alignment using
DIAMOND. Nat Methods 2015, 12(1):59–​60.
48. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification
using exact alignments. Genome Biol 2014, 15(3):R46.
49. Hyatt D, Chen GL, Locascio PF, Land ML, Larimer FW, Hauser LJ. Prodigal: pro-
karyotic gene recognition and translation initiation site identification. BMC
Bioinformatics 2010, 11:119.
50. Noguchi H, Taniguchi T, Itoh T. MetaGeneAnnotator: detecting species-​
specific patterns of ribosomal binding site for precise gene prediction in
anonymous prokaryotic and phage genomes. DNA Res 2008, 15(6):387–​396.
51. Lomsadze A, Gemayel K, Tang S, Borodovsky M. Modeling leaderless tran-
scription and atypical genes results in more accurate gene prediction in
prokaryotes. Genome Res 2018, 28(7):1079–​1089.
52. Rho M, Tang H, Ye Y. FragGeneScan: predicting genes in short and error-​prone
reads. Nucleic Acids Res 2010, 38(20):e191.
53. Kelley DR, Liu B, Delcher AL, Pop M, Salzberg SL. Gene prediction with
Glimmer for metagenomic sequences augmented by classification and
clustering. Nucleic Acids Res 2012, 40(1):e9.
54. Al-​Ajlan A, El Allali A. CNN-​ MGP: Convolutional Neural Networks for
Metagenomics Gene Prediction. Interdiscip Sci 2019, 11(4):628–​635.
55. Sommer MJ, Salzberg SL. Balrog: A universal protein model for prokaryotic
gene prediction. PLoS Comput Biol 2021, 17(2):e1008727.
56. Lowe TM, Eddy SR. tRNAscan-​ SE: a program for improved detec-
tion of transfer RNA genes in genomic sequence. Nucleic Acids Res 1997,
25(5):955–​964.
57. Laslett D, Canback B. ARAGORN, a program to detect tRNA genes and
tmRNA genes in nucleotide sequences. Nucleic Acids Res 2004, 32(1):11–​16.
58. Bland C, Ramsey TL, Sabree F, Lowe M, Brown K, Kyrpides NC, Hugenholtz
P. CRISPR recognition tool (CRT): a tool for automatic detection of clustered
regularly interspaced palindromic repeats. BMC Bioinformatics 2007, 8:209.
59. Grissa I, Vergnaud G, Pourcel C. CRISPRFinder: a web tool to identify
clustered regularly interspaced short palindromic repeats. Nucleic Acids Res
2007, 35(Web Server issue):W52–​57.
60. Biswas A, Staals RH, Morales SE, Fineran PC, Brown CM. CRISPRDetect: A
flexible algorithm to define CRISPR arrays. BMC Genomics 2016, 17:356.
61. Beghini F, McIver LJ, Blanco-​Miguez A, Dubois L, Asnicar F, Maharjan S,
Mailyan A, Manghi P, Scholz M, Thomas AM et al. Integrating taxonomic,
functional, and strain-​level profiling of diverse microbial communities with
bioBakery 3. Elife 2021, 10:e65088.
62. Milanese A, Mende DR, Paoli L, Salazar G, Ruscheweyh HJ, Cuenca M,
Hingamp P, Alves R, Costea PI, Coelho LP et al. Microbial abundance,
activity and population genomic profiling with mOTUs2. Nat Commun 2019,
10(1):1014.
360 Next-Generation Sequencing Data Analysis

63. Menzel P, Ng KL, Krogh A. Fast and sensitive taxonomic classification for
metagenomics with Kaiju. Nat Commun 2016, 7:11257.
64. Kim D, Song L, Breitwieser FP, Salzberg SL. Centrifuge: rapid and sensitive
classification of metagenomic sequences. Genome Res 2016, 26(12):1721–​1729.
65. Lu J, Breitwieser FP, Thielen P, Salzberg SL. Bracken: estimating species abun-
dance in metagenomics data. PeerJ Comput Sci 2017, 30:e104.
66. Chaumeil PA, Mussig AJ, Hugenholtz P, Parks DH. GTDB-​Tk: a toolkit to
classify genomes with the Genome Taxonomy Database. Bioinformatics 2019,
36(6):1925–​1927.
67. Parks DH, Chuvochina M, Waite DW, Rinke C, Skarshewski A, Chaumeil PA,
Hugenholtz P. A standardized bacterial taxonomy based on genome phyl-
ogeny substantially revises the tree of life. Nat Biotechnol 2018, 36(10):996–​1004.
68. Dilthey AT, Jain C, Koren S, Phillippy AM. Strain-​ level metagenomic
assignment and compositional estimation for long reads with MetaMaps. Nat
Commun 2019, 10(1):3066.
69. Fan J, Huang S, Chorlton SD. BugSeq: a highly accurate cloud platform for
long-​read metagenomic analyses. BMC Bioinformatics 2021, 22(1):160.
70. Portik DM, Brown CT, Pierce-​Ward NT. Evaluation of taxonomic classifica-
tion and profiling methods for long-​read shotgun metagenomic sequencing
datasets. BMC Bioinformatics 2022, 23(1):541.
71. UniProt C. UniProt: the universal protein knowledgebase in 2021. Nucleic
Acids Res 2021, 49(D1):D480–​D489.
72. Blum M, Chang HY, Chuguransky S, Grego T, Kandasaamy S, Mitchell A,
Nuka G, Paysan-​Lafosse T, Qureshi M, Raj S et al. The InterPro protein families
and domains database: 20 years on. Nucleic Acids Res 2021, 49(D1):D 344–​D354.
73. Galperin MY, Wolf YI, Makarova KS, Vera Alvarez R, Landsman D, Koonin
EV. COG database update: focus on microbial diversity, model organisms, and
widespread pathogens. Nucleic Acids Res 2021, 49(D1):D274–​D281.
74. Huerta-​ Cepas J, Szklarczyk D, Heller D, Hernandez-​ Plaza A, Forslund
SK, Cook H, Mende DR, Letunic I, Rattei T, Jensen LJ et al. eggNOG
5.0: a hierarchical, functionally and phylogenetically annotated orthology
resource based on 5090 organisms and 2502 viruses. Nucleic Acids Res 2019,
47(D1):D309–​D314.
75. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 2014,
30(14):2068–​2069.
76. Shaffer M, Borton MA, McGivern BB, Zayed AA, La Rosa SL, Solden LM, Liu
P, Narrowe AB, Rodriguez-​Ramos J, Bolduc B et al. DRAM for distilling micro-
bial metabolism to automate the curation of microbiome function. Nucleic
Acids Res 2020, 48(16):8883–​8900.
77. Tanizawa Y, Fujisawa T, Nakamura Y. DFAST: a flexible prokaryotic genome
annotation pipeline for faster genome publication. Bioinformatics 2018,
34(6):1037–​1039.
78. Tatusova T, DiCuccio M, Badretdin A, Chetvernin V, Nawrocki EP, Zaslavsky
L, Lomsadze A, Pruitt KD, Borodovsky M, Ostell J. NCBI prokaryotic genome
annotation pipeline. Nucleic Acids Res 2016, 44(14):6614–​6624.
79. Keegan KP, Glass EM, Meyer F. MG-​ RAST, a Metagenomics Service for
Analysis of Microbial Community Structure and Function. Methods Mol Biol
2016, 1399:207–​233.
Whole Metagenome Sequencing for Microbial Community Analysis 361

80. Chen IA, Chu K, Palaniappan K, Ratner A, Huang J, Huntemann M, Hajek P,


Ritter S, Varghese N, Seshadri R et al. The IMG/​M data management and ana-
lysis system v.6.0: new tools and advanced capabilities. Nucleic Acids Res 2021,
49(D1):D751–​D763.
81. Kanehisa M, Sato Y, Morishima K. BlastKOALA and GhostKOALA: KEGG
tools for functional characterization of genome and metagenome sequences. J
Mol Biol 2016, 428(4):726–​731.
82. Mitchell AL, Almeida A, Beracochea M, Boland M, Burgin J, Cochrane G,
Crusoe MR, Kale V, Potter SC, Richardson LJ et al. MGnify: the microbiome
analysis resource in 2020. Nucleic Acids Res 2020, 48(D1):D570–​D578.
83. Cantalapiedra CP, Hernandez-​ Plaza A, Letunic I, Bork P, Huerta-​ Cepas
J. eggNOG-​ mapper v2: functional annotation, orthology assignments,
and domain prediction at the metagenomic scale. Mol Biol Evol 2021,
38(12):5825–​5829.
84. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new
perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res
2017, 45(D1):D353–​D361.
85. Caspi R, Billington R, Keseler IM, Kothari A, Krummenacker M, Midford
PE, Ong WK, Paley S, Subhraveti P, Karp PD. The MetaCyc database of
metabolic pathways and enzymes –​a 2019 update. Nucleic Acids Res 2020,
48(D1):D445–​D453.
86. Nazeen S, Yu YW, Berger B. Carnelian uncovers hidden functional patterns
across diverse study populations from whole metagenome sequencing reads.
Genome Biol 2020, 21(1):47.
87. Ye Y, Doak TG. A parsimony approach to biological pathway reconstruc-
tion/​inference for genomes and metagenomes. PLoS Comput Biol 2009,
5(8):e1000465.
88. Seaver SMD, Liu F, Zhang Q, Jeffryes J, Faria JP, Edirisinghe JN, Mundy M,
Chia N, Noor E, Beber ME et al. The ModelSEED Biochemistry Database for
the integration of metabolic annotations and the reconstruction, comparison
and analysis of metabolic models for plants, fungi and microbes. Nucleic Acids
Res 2021, 49(D1):D1555.
89. Machado D, Andrejev S, Tramontano M, Patil KR. Fast automated reconstruc-
tion of genome-​scale metabolic models for microbial species and communi-
ties. Nucleic Acids Res 2018, 46(15):7542–​7553.
90. Paley S, Billington R, Herson J, Krummenacker M, Karp PD. Pathway tools
visualization of organism-​scale metabolic networks. Metabolites 2021, 11(2):64.
91. Capela J, Lagoa D, Rodrigues R, Cunha E, Cruz F, Barbosa A, Bastos J, Lima D,
Ferreira EC, Rocha M et al. merlin, an improved framework for the reconstruc-
tion of high-​quality genome-​scale metabolic models. Nucleic Acids Res 2022,
50(11):6052–​6066.
92. Wang H, Marcisauskas S, Sanchez BJ, Domenzain I, Hermansson D, Agren R,
Nielsen J, Kerkhoven EJ. RAVEN 2.0: a versatile toolbox for metabolic network
reconstruction and a case study on Streptomyces coelicolor. PLoS Comput Biol
2018, 14(10):e1006541.
93. Garza DR, van Verk MC, Huynen MA, Dutilh BE. Towards predicting the
environmental metabolome from metagenomics with a mechanistic model.
Nat Microbiol 2018, 3(4):456–​460.
362 Next-Generation Sequencing Data Analysis

94. Noecker C, Eng A, Srinivasan S, Theriot CM, Young VB, Jansson JK, Fredricks
DN, Borenstein E. Metabolic model-​based integration of microbiome taxo-
nomic and metabolomic profiles elucidates mechanistic links between eco-
logical and metabolic variation. mSystems 2016, 1(1):e00013–​e00015.
95. Mallick H, Franzosa EA, McLver LJ, Banerjee S, Sirota-​Madi A, Kostic AD,
Clish CB, Vlamakis H, Xavier RJ, Huttenhower C. Predictive metabolomic
profiling of microbial communities using amplicon or metagenomic
sequences. Nat Commun 2019, 10(1):3136.
96. Paulson JN, Stine OC, Bravo HC, Pop M. Differential abundance analysis for
microbial marker-​gene surveys. Nat Methods 2013, 10(12):1200–​1202.
97. McMurdie PJ, Holmes S. Waste not, want not: why rarefying microbiome
data is inadmissible. PLoS Comput Biol 2014, 10(4):e1003531.
98. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS,
Huttenhower C. Metagenomic biomarker discovery and explanation.
Genome Biol 2011, 12(6):R60.
99. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis
of taxonomic and functional profiles. Bioinformatics 2014, 30(21):3123–​3124.
100. Mandal S, Van Treuren W, White RA, Eggesbo M, Knight R, Peddada SD.
Analysis of composition of microbiomes: a novel method for studying
microbial composition. Microb Ecol Health Dis 2015, 26:27663.
101. Lin H, Peddada SD. Analysis of compositions of microbiomes with bias
correction. Nat Commun 2020, 11(1):3514.
102. Martin BD, Witten D, Willis AD. Modeling microbial abundances and
dysbiosis with beta-​binomial regression. Ann Appl Stat 2020, 14(1):94–​115.
103. Mallick H, Rahnavard A, McIver LJ, Ma S, Zhang Y, Nguyen LH, Tickle TL,
Weingart G, Ren B, Schwager EH et al. Multivariable association discovery in
population-​scale meta-​omics studies. PLoS Comput Biol 2021, 17(11):e1009442.
104. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-​Ghalith GA,
Alexander H, Alm EJ, Arumugam M, Asnicar F et al. Reproducible, inter-
active, scalable and extensible microbiome data science using QIIME 2. Nat
Biotechnol 2019, 37(8):852–​857.
105. Shi W, Qi H, Sun Q, Fan G, Liu S, Wang J, Zhu B, Liu H, Zhao F, Wang X
et al. gcMeta: a Global Catalogue of Metagenomics platform to support the
archiving, standardization and analysis of microbiome data. Nucleic Acids
Res 2019, 47(D1):D637–​D648.
363

Part IV

The Changing Landscape


of NGS Technologies and
Data Analysis
16 
What’s Next for Next-​Generation
Sequencing (NGS)?

16.1 The Changing Landscape of Next-​Generation


Sequencing (NGS)
Since its invention, massively parallel sequencing has been the major driving
force in moving life science and medicine forward. After more than a decade,
NGS continues to be the most exciting and dynamic area of genomics, and the
technology as a whole continues to evolve. As NGS is used ever more widely
in research, clinical, agricultural, and industrial applications, the drive for
technological advancements will only become greater and the competition
among existing platforms more intense. Furthermore, new sequencing tech-
nologies continue to emerge and join the fray. As a result, the landscape of
NGS is constantly changing.
Since the publication of the first edition of this book in early 2016, the gen-
eral trends in the NGS arena can be briefly summarized as follows:

1) Continued drop in sequencing cost: As of this writing (late 2022) the


sequencing cost per human genome has dropped to $300 compared to
$300,000 in 2007 when NGS started to emerge. Emerging new systems
will sequence the human genome at $100 or less.
2) Continued improvement on accuracy: Short reads technologies, such
as Illumina’s SBS, continue to reduce sequencing errors with the
use of new detection dyes, polymerases, and reaction blockers. The
advancements achieved by long-read technologies are even more sub-
stantial (see Chapter 4 for details on the PacBio and ONT platforms),
which has led to their increased adoption by the community.
3) Increased use of single DNA (or RNA) molecule sequencing: Single
molecule sequencing offers the capability to directly read individual
target DNA molecules without relying on template amplification, or
conversion to cDNA in the case of RNA.
4) Increased representation of different read lengths on different
platforms: As short and long read lengths have their strengths and also
limitations, short reads platforms such as the Illumina SBS continue to

DOI: 10.1201/9780429329180-20 365


366 Next-Generation Sequencing Data Analysis

make inroads into building long reads from short reads, while long-
read platforms have also started to provide short reads.
5) Reduction in sequencing and sample preparation time: Different
platforms use different strategies from chemistry updates, hardware
upgrades, to algorithmic improvements, to achieve quicker sequen-
cing turnaround time. To cut library prep time, constant improvements
are made to chemistries and protocols with increased amenability to
automation.
6) Decreased requirement on the amount of starting material: Historically
NGS requires large amounts of DNA or RNA to start. With the drive
to accommodate more sample types, such as those that do not gen-
erate much DNA or RNA (e.g., liquid biopsy and single cells), the
sensitivity of library making reagents and procedures has been sig-
nificantly improved.

16.2 Newer Sequencing Technologies


In Chapter 4, major NGS technologies that are widely used as the time of
writing this book are detailed. This chapter presents some of the newer tech-
nologies that are in increasing usage or in active development. Among those
in the short-read category, MGI, the instruments manufacturing arm of BGI,
has been providing a technology called DNBSEQ. Originally developed by
Complete Genomics [1], this technology is based on the use of DNA nanoball
or DNB, which is a large mass of amplified template DNA formed through
rolling circle amplification (Figure 16.1). The sequencing process is based
on the use of a procedure originally called combinatorial probe-​ anchor
ligation (or cPAL), and later refined by BGI/​MGI to become combinatorial
probe-​anchor synthesis (or cPAS) [2]. This process consists of iterative cycles
of hybridization of an anchor sequence to the template, then ligation of
fluorescence-​labeled probes, imaging for on-​board basecalling, and removal
of the anchor-​probe complex to prepare for the next cycle with a new set of
anchors and probes to interrogate the next base position. The current lineup
of MGI sequencers include DNBSEQ-​T7, DNBSEQ-​G400, and DNBSEQ-​G50,
which can produce up to 6,000 Gb, 1,440 Gb, and 150 Gb data, respectively.
The error profile of DNB sequencing is similar to that of the Illumina platform,
with the error rate slightly higher but still less than 1% [3]. Its cost per
Gb is among the lowest among currently available platforms [4]. In 2020, MGI
released a new sequencing chemistry called CoolMPS, which is based on the
use of nucleotide-​specific antibodies to detect nucleotide incorporation. This
chemistry uses unlabeled reversibly terminated nucleotides for incorporation
during synthesis, and four natural nucleotide-​ specific antibodies with
What’s Next for Next-Generation Sequencing (NGS)? 367

FIGURE 16.1
MGI/​BGI nanoball sequencing and CoolMPS chemistry. A. Nanoball sequencing starts from
circularization of DNA target molecules. After rolling circle amplification, nanoballs are formed
from circularized targets, and subsequently deposited onto a silicon-​based sequencing chip for
sequencing using the cPAS process. B. In the CoolMPS chemistry (initially called CoolNGS),
nucleotide-​specific antibodies are used for detection of incorporated nucleotides. This detection
mechanism avoids DNA “scars” derived from labeling of nucleotides with fluorescent tags,
potentially leading to increased read length. (From Gao, G., Smith, D.I. Clinical Massively
Parallel Sequencing. Clinical Chemistry, 2020, 66(1): 77–​ 88, by permission from Oxford
University Press.)

each labeled with a specific type of fluorescent dye for the detection step in
each cycle (Figure 16.1). To increase sequencing signal to background noise
ratio, multiple fluorescence dyes are attached to each antibody molecule. It
has been shown to have the potential to produce longer and more accurate
reads at lower cost [5].
Among emerging technologies, Element Biosciences offers a system based
on Avidity chemistry. Although still based on the same basic SBS process, on
the Element system the signal detection and incorporation of nucleotides are
separate, as the system does not collect sequencing signal from the nucleo-
tide incorporation process. Instead the signal is collected from binding of
nucleotides to the sequencing template. With Avidity chemistry [6], each
nucleotide attaches to a fluorescence-​emitting core, and each type of nucleo-
tide (A, C, G, or T) attaches to their own cores that emit specific fluorescence
signal for detection. The most innovative aspect of this chemistry is that
each core contains multiple fluorophores and connects to multiple copies
368 Next-Generation Sequencing Data Analysis

of a certain type of nucleotide, which enables signal amplification to boost


signal-​to-​noise ratio. After multivalent binding of the nucleotide complexes
(also called avidites, after which the sequencing chemistry is named) to the
template DNA and signal detection, the bound avidites are removed, which
is followed by a “dark” (i.e., no imaging) nucleotide incorporation step to
produce scarless DNA for a new round of avidite binding, detection, and
nucleotide incorporation. Besides the sequencing chemistry, the flow cell sur-
face where sequencing reactions take place also has ultra-​low non-​specific
binding to reduce background fluorescence and thereby further increases
signal-​to-​noise ratio. Besides reducing sequencing errors, the increased
detection sensitivity also leads to lower reagent consumption and thereby
decreased sequencing cost. Based on currently available data (as of November
2022) [6], the Element system is capable of producing 300 Gb sequencing data
with 90% of reads having quality over Q30.
Other emerging short-​ read technologies include those from Ultima,
Singular Genomics, and Omniome (part of PacBio). The Ultima system is
based on the use of (1) a new chemistry called mostly natural sequencing-​
by-​synthesis (or mnSBS); (2) an open circular wafer with a large surface for
accelerated fluidics and imaging as well as reduced reagent usage; and (3) an
AI-​based base​calling process. With the mnSBS chemistry, in each sequencing
cycle only one nucleobase is used from a reagent mix that contains mostly nat-
ural (i.e., unlabeled and non-​terminated) nucleotides and a minority (<20%)
of fluorescently labeled, non-​terminated nucleotides. This allows produc-
tion of mostly scarless DNA and at the same time a minority of fluorescently
labeled DNA molecules for imaging and base​calling. This platform has the
capability to generate billions of 300 bp reads, at an accuracy of Q30 > 85%, in
20 hours [7]. Singular Genomics provides an SBS platform based on the use
of fluorescently labeled nucleotides and 4-​color optical imaging to improve
speed and flexibility. Its first release is a G4 benchtop sequencer that produces
up to 100 Gb data with read length of up to 150 bases from each end and error
rate of <1% (mostly substitutions), in 6–​19 hours. Technical details for some
of the emerging platforms are still not available at the time of writing. Among
the currently accessible platforms, some benchmark data has become avail-
able to provide side-​by-​side comparisons on genome coverage, error rate,
alignment, and detection of various variants including SNVs, indels, and
SVs [8].
Among those in the long-​read category, there are technologies that gen-
erate native long reads similar to those from PacBio and ONT, and also
those that produce synthetic long reads. The sequencing platforms currently
being developed by Base4 and Quantapore are two examples of native long-​
reads technologies. Base4 sequencing is based on the use of a fundamentally
different process called pyrophosphorolysis. In the SBS scheme, a poly-
merase incorporates a nucleotide into an elongating DNA (or RNA) strand
and releases a pyrophosphate as a side product. With pyrophosphorolysis
sequencing, the reaction direction is reversed, i.e., the dNMP at the 3’ end
What’s Next for Next-Generation Sequencing (NGS)? 369

of a DNA strand reacts with pyrophosphate to be released from the strand


as dNTP. This sequencing-​ by-​
desynthesis process used by Base4 works
through detection of the released nucleotide [9]. The Quantapore platform
uses a nanopore-​based approach. On this platform, DNA templates need to
be labeled first with fluorophores, as instead of detecting changes in elec-
trical current as in the case of ONT, this technology detects optical signals
emitted from the fluorophores when they are activated upon passing through
the pore [10]. Other emerging native long-​read technologies are also based
on the use of nanopores, including those developed by Genia and Stratos
Genomics (now parts of Roche).
Synthetic long-read technologies are based on innovative utilization of
short-​read sequencing that links short reads together to create artificial long
reads. Technologies in this category include 10× Genomics’ linked reads [11],
MGI’s single tube Long Fragment Read (stLFR) [12], Transposase Enzyme
Linked Long-​ read Sequencing (TELL-​ seq) from Universal Sequencing
Technology [13], LoopSeq from Loop Genomics (now part of Element
Biosciences) [14], and Illumina’s Complete Long Reads. Many of these
technologies use a clonal barcoding approach, through which a large DNA
molecule is first barcoded, then shorter fragments are generated from the
large molecule to be sequenced using short-read sequencing. In this pro-
cess each short fragment generated from the same large molecule carries the
original molecule’s unique barcode. This barcode is subsequently used to
group and order short reads to reconstruct the original large DNA molecule.
Among the aforementioned synthetic long-read technologies, 10× Genomics
discontinued its linked reads offering in 2020, while Illumina has announced
Complete Long Reads in late 2022 with no technical specifics available at the
time of writing.

16.3 Continued Evolution and Growth of Bioinformatics


Tools for NGS Data Analysis
Bioinformatics tool development will continue to evolve with new sequencing
technologies as well as applications becoming available and getting adopted.
The emergence of new technologies, in turn, will lead to adaptation and evo-
lution of existing technologies. The increased change in read length and other
aspects of sequencing output, such as diversity of sequencing error models
form different platforms, will lead to development of new tools and revi-
sion of existing tools. As higher read length increases sequence information
content and uniqueness, which in turn leads to increased “assemblability”
or “mappability,” new alignment algorithms or updated versions of existing
ones will continue to be developed to harness the power afforded by this
370 Next-Generation Sequencing Data Analysis

increase in read length. For example, SPAdes, originally developed for de


novo assembly of Illumina short reads [15], has been revised to enable use
of both short and long but less accurate PacBio and ONT reads to achieve
hybrid assembly [16]. New long-​read de novo genome assemblers, such as
miniasm [17], Flye [18], NECAT [19], and Raven [20], have been developed
more recently. To accommodate mapping of long reads to a reference genome,
BWA-​MEM was a first attempt to align long reads with the widely used BWA
algorithm. With ultra-​long but noisier reads becoming more common, add-
itional aligners, such as minimap [17] and subsequently minimap2 [21], are
developed and updated constantly [22] with rapid advancements in sequen-
cing technologies.
Significantly longer reads associated with third-​or future-​ generation
sequencing technologies, as well as synthetic long reads, will not only
improve de novo genome assembly and mapping to reference sequences but
also all other NGS applications. For example, increased read length in RNA-​
seq can lead to recognition of different transcripts that are produced from the
same gene, and therefore facilitate studies of alternative splicing. Algorithms
and tools for other applications or steps, from basecalling, variant calling,
ChIP-​seq peaking calling, to DNA methylation sequencing, and metagenome
characterization, are under constant development. While new ones are
continuously being introduced, many existing algorithms and tools are
also under constant revision. As basecalling is highly platform-​dependent,
basecallers are usually developed as part of the sequencing platform devel-
opment process. While there are also third-​party basecallers being developed
in an attempt to further improve performance, efforts on algorithmic and
software tool development are mostly focused on downstream analyses.
To help illustrate how data analytic tool development closely follows the
development of new technologies and applications, Figure 16.2 shows the
total number of publications on scRNA-​seq data analysis algorithms every
year from 2013 to 2021. As a new application, single-cell RNA-​seq has seen
continuous growth in algorithmic development and utilization since 2013.
While the numbers do not directly measure the total number of new or
updated scRNA-​seq algorithms, they do to a large degree reflect the amount
of algorithmic development efforts as well as the demand in this direction.
Algorithmic development and utilization in other new applications and tech-
nologies show a similar trend.

16.4 Efficient Management of NGS Analytic Workflows


With the active development of algorithms and the wide array of bioinfor-
matic tools becoming available, efficient workflow management
What’s Next for Next-Generation Sequencing (NGS)? 371

FIGURE 16.2
The increase in the number of single-cell RNA-​seq algorithm-​related publications from 2013 till
2021. (Data source: Google Scholar, using “single cell RNA-​seq” AND (algorithm OR method OR
tool) as query term.)

has become increasingly important. The clinical use of NGS on diagnosis,


genetic risk assessment, and patient management places further
demands on NGS data workflow management. The quest for increased
reproducibility is another impetus for effective workflow management,
as the tool(s) used, their version, and their configuration all have effects
on final results. Commercial software systems, such as CLC Genomics
Workbench, DNAnexus, DNAstar, PierianDx, QIAGEN Clinical Insight
Interpret, Seven Bridges, and SciDAP, tend to streamline their analytical
workflows in one package. Because bioinformatic tools developed
out of academic settings tend to be more specialized and as a result a bit
fragmented, effective workflow management strategies and systems are
becoming increasingly needed. As a result, usage of workflow manage-
ment systems has been on the rise to overcome the limitations of using
the traditional way of downloading and deploying tools one by one for
a workflow. Such limitations involve the difficulty to deploy associated
system libraries (including dependencies), compilers or interpreters of
different versions, for different computing environments. Several key tech-
nologies in computer science, including the use of containers, workflow
description languages, and workflow engines, have been adopted for man-
aging NGS analytic workflows.
372 Next-Generation Sequencing Data Analysis

In computer science terms, a container is an executable unit of software


package that contains everything an application needs to run, including
all code and requisite libraries and dependencies. This technology allows
the software in a container to run identically in different computing envir-
onments, overcoming the issues associated with running the same tool on
different platforms. Two widely used container engines in the bioinformatics
arena are Docker and Singularity. Community efforts such as Biocontainers
[23] provide pre-​built bioinformatics software containers.
A workflow description language enables a user to describe a work-
flow that can be executed on different platforms, but without the need for
platform-​specific execution details. Commonly used workflow descrip-
tion languages include the Common Workflow Language or CWL, and the
Workflow Description Language or WDL. With the use of such workflow
description languages, the description of a workflow becomes separate from
the physical computing platform that runs it. A workflow engine, also called
workflow management system, determines how a workflow is executed and
controlled under different computing environments. Workflow engines in
the NGS field include Galaxy [24], Cromwell as used in Terra [25], Nextflow
[26], and CWL-​Airflow [27]. These workflow management systems enable
seamless execution of tools assembled into an integrated pipeline (e.g., RNA-​
seq quantification followed by differential expression analysis), as well as
provide infrastructure and guidelines for management and distribution of
NGS and other bioinformatics packages. With increased use of containers
and workflow management systems, NGS data analysis will become more
streamlined and standardized, thereby improving reproducibility, as well as
portability (for porting workflows across different computing environments)
and scalability (for efficient use of large-​scale computational resources).

16.5 Deepening Applications of NGS to Single-Cell


and Spatial Sequencing
Among the various new directions that NGS has empowered and grown into
is single-cell sequencing. In this revision, a new chapter is dedicated to single-
cell transcriptomics. Beyond transcriptomic information, information on
other -​omic aspects of single cells is also required to reveal their inner working
mechanisms and the tremendous heterogeneity among them. NGS has been
making inroads into these aspects, with examples from single-cell ATAC-​seq,
single-cell whole genome/​exome sequencing, and single-cell epigenomics.
Single-cell ATAC-​seq, or Assay for Transposase-​Accessible Chromatin using
sequencing, is a technique to reveal chromatin accessibility landscape for
transcription control. Available exemplary studies have demonstrated the
What’s Next for Next-Generation Sequencing (NGS)? 373

rich information it can provide for analysis of gene regulatory programs [28].
Compared to tools developed for scRNA-​seq, scATAC-​seq data analysis tools
are still low in numbers and not as well developed. Currently available tools
include SnapATAC [29], cisTopic [30], SCALE [31], and Signac [32].
Single-cell genome/​exome sequencing, with the goal of tracking som-
atic evolution and revealing genetic heterogeneity at the single-cell level,
has been made possible with whole genome amplification methods. These
methods include multiple displacement amplification (MDA) [33], multiple
annealing and loop-​based amplification cycles (MALBAC) [34], degenerate
oligonucleotide-​primed PCR (DOP-​PCR) [35], or commercially available kits
such as PicoPLEX and RepliG [36]. As each diploid cell only has two sets of
chromosomes and therefore a very low amount of DNA (~6.5 pg in a typical
human cell), the coverage of whole genome amplification is typically uneven
across the genome due to stochastic effects, amplification errors, and locus-​
specific amplification bias. Such issues have prevented scaling-​up of single-​
cell genomics to a level that can be comparable to single-cell transcriptomics.
Progress has been made to overcome these issues with the development of
emulsion MDA (or eMDA) [37], single droplet MDA (or sd-​MDA) [38], and
direct library preparation (DLP) [39]. To call SNVs from single-cell genomics
data, currently a relatively short list of tools is available, including SCcaller
[40], SCAN-​SNV [41], and Single Cell Genotyper [42]. SNV Calling can also
be made from scRNA-​seq data, or coupled scDNA-​seq and scRNA-​seq data,
using tools such as SSrGE [43]. Tools for calling CNVs or structural variants
include AneuFinder [44] and Ginkgo [45].
Single-cell epigenomics offers another dimension for single-cell sequencing.
Strategies such as single-cell whole genome bisulfite sequencing (scWGBS)
[46], single-cell reduced representation bisulfite sequencing (scRRBS) [47],
and single-​nucleus methylome sequencing ver2 (snmC-​seq2) [48] have been
used to detect DNA methylation as a epigenetic marker for cell typing. Tools
such as scBS-​map [49] can be used for reads alignment, and Methylpy [50]
for calling of unmethylated and methylated cytosines. Simultaneous inter-
rogation of both the epigenome and genome of single cells has also been
made possible with methods such as epi-​gSCAR [51]. Compared to scRNA-​
seq and scATAC-​seq conducted on high-​throughput platforms such as 10×
Chromium, the throughput of single-cell genome and epigenome sequencing
is still lower, although this may well change over time.
While single-cell sequencing offers unprecedented resolution, isolation of
single cells (see Chapter 8) typically leads to the loss of contextual informa-
tion about their original location in their native tissue microenvironment.
Investigation into such spatial information provides insights on regional
specificity and cross-​region heterogeneity, e.g., when comparing a patho-
genic region with the surrounding normal region on the same slide. Spatial
transcriptomics, enabled by rapid technology development from both aca-
demia and industry [52], is increasingly used to provide this additional layer
374 Next-Generation Sequencing Data Analysis

of information. While this new field is still rapidly evolving, a number of


platforms have been gradually adopted by the community, included among
which are academically developed platforms such as Slide-​seqV2 [53], and
commercial ones such as Visium from 10× Genomics, GeoMx Digital Spatial
Profiler from Nanostring, and MERFISH from Vizgen. These systems, based
on a diversity of detection mechanisms, have different technical capabilities
in terms of spatial resolution, physical dimensions of the area to be assayed,
and transcriptomic coverage (whole transcriptome vs. targeted genes). To
analyze data from these systems, there are currently some tools available,
such as Space Ranger from 10× Genomics, Giotto [54], and BayesSpace
[55], to help identify cell types, characterize gene expression profiles and
infer cell-​cell interactions in a spatial context. To meet the challenges in spa-
tial transcriptomics towards the goal of integrating gene expression pro-
file, spatial context, and cell morphology for deeper cell typing, more tools
are needed. More comprehensive tools are also needed to integrate spatial
transcriptomics data, with other omics data including spatial proteomics and
metabolomics.

16.6 Increasing Use of Machine Learning


in NGS Data Analytics
As a branch of artificial intelligence, machine learning (ML) trains algorithms
to uncover patterns in large data sets and thereby makes classifications or
predictions without being explicitly programmed. Within ML, deep learning
(DL) with the use of neural networks is well suited for tasks that involve
large volumes of highly complex and information-​rich data. As NGS has
become a major producer of big data, besides astronomy, YouTube, and social
media, application of what ML and DL can offer is warranted [56, 57]. In
earlier chapters, such as Chapters 8 and 11, the applications of ML and DL to
single-cell sequencing data analysis and detection of clinically relevant gen-
etic variants have been covered. Besides these areas, nearly all aspects of NGS
data analysis have seen increased use of ML and DL. Read mapping and
variant calling, the two most basic steps, have been greatly improved by the
application of ML and DL approaches. In the precisionFDA Truth Challenge
V2, the majority of read mapping and variant calling pipelines employ ML
and DL approaches [58]. The Illumina DRAGEN reads mapper +​variant
caller system, for example, applies graph-​based mapping and supervised ML-​
based variant calling to improve accuracy as well as speed. For the mapping
step, DRAGEN uses phased graph reference genome built through stitching
alternate haplotypes into the standard reference genome, and thereby cre-
ating alternative graph paths for seed mapping and alignment of reads. Such
a reference genome based on the use of graph representation, a subfield
What’s Next for Next-Generation Sequencing (NGS)? 375

of ML, represents more accurately the polymorphic nature of a species’ or


population’s genome. In practice, this alt-​aware approach leads to improved
mapping performance by reducing ambiguity when reads containing alter-
nate haplotypes are mapped more accurately than using the standard refer-
ence genome alone. For the variant calling step, DRAGEN uses a supervised
ML model trained to incorporate read-​based and contextual features that
is then employed to recalibrate the QUAL and GQ fields from a standard
VCF output to increase variant calling accuracy. Among other examples,
the Seven Bridges GRAF pipeline also uses a pan-​genome graph reference,
created by incorporating known alternate haplotypes and integrating gen-
omic polymorphic information from multiple variant databases, to improve
reads alignment. To improve variant calling, the pipeline uses an adaptive
ML model to remove false-​positive variants. Similarly Sentieon’s DNAscope
and TNscope pipelines also use ML-​enhanced variant filtering to improve
the accuracy of calling germline and somatic variants, respectively.
DeepVariant, an open-​source variant caller, is the first variant caller among
all variant callers that uses a DL-​based framework. Originally developed for
classifying images [59], this Tensorflow-​based framework uses a convolu-
tional neural network (CNN) for calling small germline variants from short
reads. Since its introduction, DL-​based methods have grown significantly
overtaking statistical model-​based methods. Open-​source tools for calling
variants from long reads, such as those generated from ONT and PacBio,
have been dominated by ML/​ DL-​
based methods since the beginning,
largely out of the need to deal with the higher sequencing error rates on these
platforms. The PEPPER-​Margin-​DeepVariant pipeline, for example, uses a
recurrent neural network (RNN) for variant filtering and HMM for phasing
and haplotyping, prior to variant calling by DeepVariant [60]. Examples of
other such tools include Clair/​Clair3 based on RNNs [61, 62], NanoCaller
on deep CNN [63], and Longshot on pair-​HMM [64]. For structural variant
calling, the use of DL has not been as wide at the time of writing, but existing
tools such as DeepSV [65] and dudeML [66] have shown great promise.
Besides read mapping and variant calling, many of the NGS applications
covered in this book have also seen increasing use of ML and DL. For
example, in performing bulk RNA-​seq, statistical models are usually used to
identify differentially expressed genes. Emerging ML-​based methods have
been shown to be able to identify DEGs, some of which may be missed by
statistical methods. For example, an ML model based on the use of InfoGain
feature selection and Logistic Regression classification was found to be
robust and have improved sensitivity [67]. One benefit of ML-​based gene
feature selections is that they do not make assumptions on the distribution of
gene transcript counts. In ChIP-​seq, peak detection is a major step to reveal
regions of the genome where mapped reads are enriched that indicate epi-
genetic interactions. As covered in Chapter 13, peaks are typically called
using statistical methods. To overcome the challenges these methods face,
such as uneven background noise and high false-​positive rates, DL-​based
376 Next-Generation Sequencing Data Analysis

approaches have been employed. For example, a CNN-​ based pipeline


called CNN-​Peaks uses partially labeled peaks inspected by researchers and
genome annotations as input to first train a model for subsequent peak detec-
tion. This supervised learning process takes into consideration variations in
local context and background noise and sets specific peak detection cut-​off
values accordingly for each genomic region to reduce false-​positive rates. In
methyl-​seq, currently available ML/​DL-​based methods are mostly designed
to detect epigenetic base modifications from data generated on long-​read
platforms, such as DeepMP [68] and DeepSignal-​plant [69] for ONT data,
and a holistic kinetic (HK) model for PacBio data [70].
Beyond the various NGS steps and applications, ML and DL can also
greatly facilitate integration of data collected from multiple -​omics technolo-
gies, including genomics, transcriptomics, epigenomics, proteomics, and
metabolomics. Data collected using these -​omics technologies informs on the
different layers of a biological system, and integration of such complementary
information leads to a more holistic view of the system. Towards this direc-
tion, beyond basic research ML, especially DL, has been accelerating clinical
application of multi-​omics data to disease subtyping, biomarker discovery,
disease prognosis, treatment outcome prediction, and drug repurposing. For
example, to predict severity of pediatric irritable bowel syndrome (IBS), a
Random Forest-​based classifier is developed from integration of shotgun
metagenomics and metabolomics data [71]. This classifier is optimized based
on the selection and use of key features derived from the metagenomics and
metabolomics data, including microbial diversity, functional pathways, and
fecal metabolites. Through the use of such gut microbiome and metabolites
features as integrated diagnostic biomarkers, the classifier can reliably pre-
dict IBS status and thereby help to stratify patients for improved disease man-
agement. As another example, an oncology molecular classifier integrates
genome and transcriptome sequencing data to identify tissue-​of-​origin (TOO)
for cancers of unknown primary (or CUP), a syndrome of metastatic cancers
with no clinically detectable primary sites of origin. Without knowing their
TOO, CUP has very poor prognosis. By combining DNA-​level mutation and
RNA-​level gene expression information, the DL model makes predictions on
tumor TOO across a broad spectrum of tumor types at high accuracy, thereby
providing diagnostic utility and guiding therapeutic intervention [72].

References
1. Drmanac R, Sparks AB, Callow MJ, Halpern AL, Burns NL, Kermani BG,
Carnevali P, Nazarenko I, Nilsen GB, Yeung G et al. Human genome sequen-
cing using unchained base reads on self-​assembling DNA nanoarrays. Science
2010, 327(5961):78–​81.
What’s Next for Next-Generation Sequencing (NGS)? 377

2. Fehlmann T, Reinheimer S, Geng C, Su X, Drmanac S, Alexeev A, Zhang C,


Backes C, Ludwig N, Hart M et al. cPAS-​based sequencing on the BGISEQ-​500
to explore small non-​coding RNAs. Clin Epigenetics 2016, 8:123.
3. Zhu K, Du P, Xiong J, Ren X, Sun C, Tao Y, Ding Y, Xu Y, Meng H, Wang
CC et al. Comparative Performance of the MGISEQ-​2000 and Illumina X-​Ten
Sequencing Platforms for Paleogenomics. Front Genet 2021, 12:745508.
4. Tedersoo L, Albertsen M, Anslan S, Callahan B. Perspectives and Benefits of
High-​Throughput Long-​Read Sequencing in Microbial Ecology. Appl Environ
Microbiol 2021, 87(17):e0062621.
5. Drmanac S, Callow M, Chen L, Zhou P, Eckhardt L, Xu C, Gong M, Gablenz
S, Rajagopal J, Yang Q et al. CoolMPS™: Advanced massively parallel sequen-
cing using antibodies specific to each natural nucleobase. bioRxiv 2020,
doi: https://​doi.org/​10.1101/​2020.02.19.953​307
6. Arslan S, Garcia FJ, Guo M, Kellinger MW, Kruglyak S, LeVieux JA, Mah
AH, Wang H, Zhao J, Zhou C. Sequencing by avidity enables high accuracy
with low reagent consumption. bioRxiv 2022, doi: https://​doi.org/​10.1101/​
2022.11.03.514​117
7. Almogy G, Pratt M, Oberstrass F, Lee L, Mazur D, Beckett N, Barad O, Soifer
I, Perelman E, Etzioni Y et al. Cost-​efficient whole genome-​sequencing using
novel mostly natural sequencing-​by-​synthesis chemistry and open fluidics
platform. bioRxiv 2022, doi: https://​doi.org/​10.1101/​2022.05.29.493​900
8. Foox J, Tighe SW, Nicolet CM, Zook JM, Byrska-​Bishop M, Clarke WE, Khayat
MM, Mahmoud M, Laaguiby PK, Herbert ZT et al. Performance assessment of
DNA sequencing platforms in the ABRF Next-​Generation Sequencing Study.
Nat Biotechnol 2021, 39(9):1129–​1140.
9. Puchtler TJ, Johnson K, Palmer RN, Talbot EL, Ibbotson LA, Powalowska PK,
Knox R, Shibahara A, P MSC, Newell OJ et al. Single-​molecule DNA sequen-
cing of widely varying GC-​content using nucleotide release, capture and
detection in microdroplets. Nucleic Acids Res 2020, 48(22):e132.
10. Marx V. Nanopores: a sequencer in your backpack. Nat Methods 2015,
12(11):1015–​1018.
11. Zheng GX, Lau BT, Schnall-​ Levin M, Jarosz M, Bell JM, Hindson CM,
Kyriazopoulou-​Panagiotopoulou S, Masquelier DA, Merrill L, Terry JM et al.
Haplotyping germline and cancer genomes with high-​throughput linked-​read
sequencing. Nat Biotechnol 2016, 34(3):303–​311.
12. Wang O, Chin R, Cheng X, Wu MKY, Mao Q, Tang J, Sun Y, Anderson E, Lam
HK, Chen D et al. Efficient and unique cobarcoding of second-​generation
sequencing reads from long DNA molecules enabling cost-​ effective and
accurate sequencing, haplotyping, and de novo assembly. Genome Res 2019,
29(5):798–​808.
13. Chen Z, Pham L, Wu TC, Mo G, Xia Y, Chang PL, Porter D, Phan T, Che H,
Tran H et al. Ultralow-​input single-​tube linked-​read library method enables
short-​read second-​ generation sequencing systems to routinely generate
highly accurate and economical long-​range sequencing information. Genome
Res 2020, 30(6):898–​909.
14. Liu S, Wu I, Yu YP, Balamotis M, Ren B, Ben Yehezkel T, Luo JH. Targeted
transcriptome analysis using synthetic long read sequencing uncovers
isoform reprograming in the progression of colon cancer. Commun Biol 2021,
4(1):506.
378 Next-Generation Sequencing Data Analysis

15. Nurk S, Bankevich A, Antipov D, Gurevich AA, Korobeynikov A, Lapidus A,


Prjibelski AD, Pyshkin A, Sirotkin A, Sirotkin Y et al. Assembling single-​cell
genomes and mini-​metagenomes from chimeric MDA products. J Comput Biol
2013, 20(10):714–​737.
16. Antipov D, Korobeynikov A, McLean JS, Pevzner PA. hybridSPAdes: an
algorithm for hybrid assembly of short and long reads. Bioinformatics 2016,
32(7):1009–​1015.
17. Li H. Minimap and miniasm: fast mapping and de novo assembly for noisy
long sequences. Bioinformatics 2016, 32(14):2103–​2110.
18. Kolmogorov M, Yuan J, Lin Y, Pevzner PA. Assembly of long, error-​prone
reads using repeat graphs. Nat Biotechnol 2019, 37(5):540–​546.
19. Chen Y, Nie F, Xie SQ, Zheng YF, Dai Q, Bray T, Wang YX, Xing JF, Huang ZJ,
Wang DP et al. Efficient assembly of nanopore reads via highly accurate and
intact error correction. Nat Commun 2021, 12(1):60.
20. Vaser R, Šikić M. Time-​and memory-​efficient genome assembly with Raven.
Nat Comput Sci 2021, 1(5):332–​336.
21. Li H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics
2018, 34(18):3094–​3100.
22. Li H. New strategies to improve minimap2 alignment accuracy. Bioinformatics
2021, 37(23):4572–​4574.
23. Bai J, Bandla C, Guo J, Vera Alvarez R, Bai M, Vizcaino JA, Moreno P, Gruning
B, Sallou O, Perez-​Riverol Y. BioContainers Registry: Searching Bioinformatics
and Proteomics Tools, Packages, and Containers. J Proteome Res 2021,
20(4):2056–​2061.
24. Jalili V, Afgan E, Gu Q, Clements D, Blankenberg D, Goecks J, Taylor J,
Nekrutenko A. The Galaxy platform for accessible, reproducible and col-
laborative biomedical analyses: 2020 update. Nucleic Acids Res 2020,
48(W1):W395–​W402.
25. Terra (https://​app.terra.bio/​)
26. Di Tommaso P, Chatzou M, Floden EW, Barja PP, Palumbo E, Notredame C.
Nextflow enables reproducible computational workflows. Nat Biotechnol 2017,
35(4):316–​319.
27. Kotliar M, Kartashov AV, Barski A. CWL-​Airflow: a lightweight pipeline man-
ager supporting Common Workflow Language. GigaScience 2019, 8(7):giz084.
28. Zhang K, Hocker JD, Miller M, Hou X, Chiou J, Poirion OB, Qiu Y, Li YE,
Gaulton KJ, Wang A et al. A single-​cell atlas of chromatin accessibility in the
human genome. Cell 2021, 184(24):5985–​6001 e5919.
29. Fang R, Preissl S, Li Y, Hou X, Lucero J, Wang X, Motamedi A, Shiau AK,
Zhou X, Xie F et al. Comprehensive analysis of single cell ATAC-​seq data with
SnapATAC. Nat Commun 2021, 12(1):1337.
30. Bravo Gonzalez-​Blas C, Minnoye L, Papasokrati D, Aibar S, Hulselmans G,
Christiaens V, Davie K, Wouters J, Aerts S. cisTopic: cis-​regulatory topic mod-
eling on single-​cell ATAC-​seq data. Nat Methods 2019, 16(5):397–​400.
31. Xiong L, Xu K, Tian K, Shao Y, Tang L, Gao G, Zhang M, Jiang T, Zhang QC.
SCALE method for single-​cell ATAC-​seq analysis via latent feature extraction.
Nat Commun 2019, 10(1):4576.
32. Stuart T, Srivastava A, Madad S, Lareau CA, Satija R. Single-​cell chromatin
state analysis with Signac. Nat Methods 2021, 18(11):1333–​1341.
What’s Next for Next-Generation Sequencing (NGS)? 379

33. Dean FB, Nelson JR, Giesler TL, Lasken RS. Rapid amplification of plasmid
and phage DNA using Phi 29 DNA polymerase and multiply-​primed rolling
circle amplification. Genome Res 2001, 11(6):1095–​1099.
34. Zong C, Lu S, Chapman AR, Xie XS. Genome-​ wide detection of single-​
nucleotide and copy-​number variations of a single human cell. Science 2012,
338(6114):1622–​1626.
35. Telenius H, Carter NP, Bebb CE, Nordenskjold M, Ponder BA, Tunnacliffe A.
Degenerate oligonucleotide-​primed PCR: general amplification of target DNA
by a single degenerate primer. Genomics 1992, 13(3):718–​725.
36. Imamura H, Monsieurs P, Jara M, Sanders M, Maes I, Vanaerschot M,
Berriman M, Cotton JA, Dujardin JC, Domagalska MA. Evaluation of whole
genome amplification and bioinformatic methods for the characterization of
Leishmania genomes at a single cell level. Sci Rep 2020, 10(1):15043.
37. Fu Y, Zhang F, Zhang X, Yin J, Du M, Jiang M, Liu L, Li J, Huang Y, Wang J.
High-​throughput single-​cell whole-​genome amplification through centrifugal
emulsification and eMDA. Commun Biol 2019, 2:147.
38. Hosokawa M, Nishikawa Y, Kogawa M, Takeyama H. Massively parallel whole
genome amplification for single-​cell sequencing using droplet microfluidics.
Sci Rep 2017, 7(1):5199.
39. Zahn H, Steif A, Laks E, Eirew P, VanInsberghe M, Shah SP, Aparicio S,
Hansen CL. Scalable whole-​genome single-​cell library preparation without
preamplification. Nat Methods 2017, 14(2):167–​173.
40. Dong X, Zhang L, Milholland B, Lee M, Maslov AY, Wang T, Vijg J. Accurate
identification of single-​nucleotide variants in whole-​genome-​amplified single
cells. Nat Methods 2017, 14(5):491–​493.
41. Luquette LJ, Bohrson CL, Sherman MA, Park PJ. Identification of somatic
mutations in single cell DNA-​seq using a spatial model of allelic imbalance.
Nat Commun 2019, 10(1):3908.
42. Roth A, McPherson A, Laks E, Biele J, Yap D, Wan A, Smith MA, Nielsen
CB, McAlpine JN, Aparicio S et al. Clonal genotype and population struc-
ture inference from single-​ cell tumor sequencing. Nat Methods 2016,
13(7):573–​576.
43. Poirion O, Zhu X, Ching T, Garmire LX. Using single nucleotide variations
in single-​cell RNA-​seq to identify subpopulations and genotype-​phenotype
linkage. Nat Commun 2018, 9(1):4892.
44. Bakker B, Taudt A, Belderbos ME, Porubsky D, Spierings DC, de Jong TV,
Halsema N, Kazemier HG, Hoekstra-​Wakker K, Bradley A et al. Single-​cell
sequencing reveals karyotype heterogeneity in murine and human malignan-
cies. Genome Biol 2016, 17(1):115.
45. Garvin T, Aboukhalil R, Kendall J, Baslan T, Atwal GS, Hicks J, Wigler M,
Schatz MC. Interactive analysis and assessment of single-​cell copy-​number
variations. Nat Methods 2015, 12(11):1058–​1060.
46. Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J, Andrews
SR, Stegle O, Reik W, Kelsey G. Single-​cell genome-​wide bisulfite sequencing
for assessing epigenetic heterogeneity. Nat Methods 2014, 11(8):817–​820.
47. Guo H, Zhu P, Guo F, Li X, Wu X, Fan X, Wen L, Tang F. Profiling DNA
methylome landscapes of mammalian cells with single-​ cell reduced-​
representation bisulfite sequencing. Nat Protoc 2015, 10(5):645–​659.
380 Next-Generation Sequencing Data Analysis

48. Luo C, Rivkin A, Zhou J, Sandoval JP, Kurihara L, Lucero J, Castanon R, Nery
JR, Pinto-​Duarte A, Bui B et al. Robust single-​cell DNA methylome profiling
with snmC-​seq2. Nat Commun 2018, 9(1):3824.
49. Wu P, Gao Y, Guo W, Zhu P. Using local alignment to enhance single-​cell
bisulfite sequencing data efficiency. Bioinformatics 2019, 35(18):3273–​3278.
50. Schultz MD, He Y, Whitaker JW, Hariharan M, Mukamel EA, Leung D,
Rajagopal N, Nery JR, Urich MA, Chen H et al. Human body epigenome
maps reveal noncanonical DNA methylation variation. Nature 2015,
523(7559):212–​216.
51. Niemoller C, Wehrle J, Riba J, Claus R, Renz N, Rhein J, Bleul S, Stosch JM,
Duyster J, Plass C et al. Bisulfite-​free epigenomics and genomics of single cells
through methylation-​sensitive restriction. Commun Biol 2021, 4(1):153.
52. Liao J, Lu X, Shao X, Zhu L, Fan X. Uncovering an organ’s molecular archi-
tecture at single-​cell resolution by spatially resolved transcriptomics. Trends
Biotechnol 2021, 39(1):43–​58.
53. Stickels RR, Murray E, Kumar P, Li J, Marshall JL, Di Bella DJ, Arlotta P,
Macosko EZ, Chen F. Highly sensitive spatial transcriptomics at near-​cellular
resolution with Slide-​seqV2. Nat Biotechnol 2021, 39(3):313–​319.
54. Dries R, Zhu Q, Dong R, Linus Eng C-​H, Li H, Liu K, Fu Y, Zhao T, Sarkar A,
Bao F et al. Giotto: a toolbox for integrative analysis and visualization of spa-
tial expression data. Genome Biol 2021, 22(1):78.
55. Zhao E, Stone MR, Ren X, Guenthoer J, Smythe KS, Pulliam T, Williams SR,
Uytingco CR, Taylor SEB, Nghiem P et al. Spatial transcriptomics at subspot
resolution with BayesSpace. Nat Biotechnol 2021, 39(11):1375–​1384.
56. Stephens ZD, Lee SY, Faghri F, Campbell RH, Zhai CX, Efron MJ, Iyer R, Schatz
MC, Sinha S, Robinson GE. Big Data: Astronomical or Genomical? PLoS Biol
2015, 13(7).
57. Schmidt B, Hildebrandt A. Next-​generation sequencing: big data meets high
performance computing. Drug Discov Today 2017, 22(4):712–​717.
58. Olson ND, Wagner J, McDaniel J, Stephens SH, Westreich ST, Prasanna AG,
Johanson E, Boja E, Maier EJ, Serang O et al. PrecisionFDA Truth Challenge
V2: Calling variants from short-​and long-​reads in difficult-​to-​map regions.
Cell Genom 2022, 2(5):100129.
59. Poplin R, Chang PC, Alexander D, Schwartz S, Colthurst T, Ku A, Newburger
D, Dijamco J, Nguyen N, Afshar PT et al. A universal SNP and small-​indel
variant caller using deep neural networks. Nat Biotechnol 2018, 36(10):983–​987.
60. Shafin K, Pesout T, Chang PC, Nattestad M, Kolesnikov A, Goel S, Baid G,
Kolmogorov M, Eizenga JM, Miga KH et al. Haplotype-​aware variant calling
with PEPPER-​Margin-​DeepVariant enables high accuracy in nanopore long-​
reads. Nat Methods 2021, 18(11):1322–​1332.
61. Luo R, Wong C-​L, Wong Y-​S, Tang C-​I, Liu C-​M, Leung C-​M, Lam T-​W.
Exploring the limit of using a deep neural network on pileup data for germline
variant calling. Nat Mach Intell 2020, 2(4):220–​227.
62. Zheng Z, Li S, Su J, Leung AW-​S, Lam T-​W, Luo R. Symphonizing pileup and
full-​alignment for deep learning-​based long-​read variant calling. Nat Comput
Sci 2021, 2(12):797–​803.
63. Ahsan MU, Liu Q, Fang L, Wang K. NanoCaller for accurate detection of
SNPs and indels in difficult-​to-​map regions from long-​read sequencing by
haplotype-​aware deep neural networks. Genome Biol 2021, 22(1):261.
What’s Next for Next-Generation Sequencing (NGS)? 381

64. Edge P, Bansal V. Longshot enables accurate variant calling in diploid genomes
from single-​molecule long read sequencing. Nat Commun 2019, 10(1):4660.
65. Cai L, Wu Y, Gao J. DeepSV: accurate calling of genomic deletions from high-​
throughput sequencing data using deep convolutional neural network. BMC
Bioinformatics 2019, 20(1):665.
66. Hill T, Unckless RL. A Deep Learning Approach for Detecting Copy Number
Variation in Next-​Generation Sequencing Data. G3 2019, 9(11):3575–​3582.
67. Wang L, Xi Y, Sung S, Qiao H. RNA-​seq assistant: machine learning based
methods to identify more transcriptional regulated genes. BMC Genomics
2018, 19(1):546.
68. Bonet J, Chen M, Dabad M, Heath S, Gonzalez-​Perez A, Lopez-​Bigas N,
Lagergren J. DeepMP: a deep learning tool to detect DNA base modifications
on Nanopore sequencing data. Bioinformatics 2022, 38(5):1235–​1243.
69. Ni P, Huang N, Nie F, Zhang J, Zhang Z, Wu B, Bai L, Liu W, Xiao CL, Luo F
et al. Genome-​wide detection of cytosine methylations in plant from Nanopore
data using deep learning. Nat Commun 2021, 12(1):5976.
70. Tse OYO, Jiang P, Cheng SH, Peng W, Shang H, Wong J, Chan SL, Poon LCY,
Leung TY, Chan KCA et al. Genome-​wide detection of cytosine methyla-
tion by single molecule real-​time sequencing. Proc Natl Acad Sci U S A 2021,
118(5):e2019768118.
71. Hollister EB, Oezguen N, Chumpitazi BP, Luna RA, Weidler EM, Rubio-​
Gonzales M, Dahdouli M, Cope JL, Mistretta TA, Raza S et al. Leveraging
human microbiome features to diagnose and stratify children with irritable
bowel syndrome. J Mol Diagn 2019, 21(3):449–​461.
72. Abraham J, Heimberger AB, Marshall J, Heath E, Drabick J, Helmstetter A, Xiu
J, Magee D, Stafford P, Nabhan C et al. Machine learning analysis using 77,044
genomic and transcriptomic profiles to accurately predict tumor type. Transl
Oncol 2021, 14(3):101016.
383

Appendix I
Common File Types Used in NGS
Data Analysis

BAM: A file format for storing reads alignment data. It is the binary version
of the SAM format (see below). Compared to its equivalent SAM file, a
BAM file is considerably smaller in size and much faster to load. Unlike
SAM files, however, the BAM format is not human-​readable. BAM files
have a file extension of .bam. Some tools require BAM files to be indexed.
Besides the .bam file, an indexed BAM file also has a companion index
file of the same name but with a different file extension (.bai).
BCF: Binary VCF (see VCF). While it is equivalent to VCF, BCF is much
smaller in file size due to compression, and therefore achieves high effi-
ciency in file transfer and parsing.
BCL: Binary basecall files generated from Illumina’s proprietary basecalling
process.
BED: Browser Extensible Display format used to describe genes or other
genomic features in a genome browser. It is a tab-​delimited text format
that defines how genes or genomic features are displayed as an anno-
tation track in a genome browser such as the UCSC Genome Browser.
Each entry line contains three mandatory fields (chrom, chromStart, and
chromEnd, specifying for each genomic feature the particular chromo-
some it is located on and the start and end coordinates) and nine optional
fields. Binary PED files (see below) are also referred to as BED files, but
this is a totally different file format.
bedGraph: Similar to the BED format, bedGraph provides descriptions of
genomic features for their display in a genome browser. Distinctively
the bedGraph format allows display of continuous values, such as prob-
ability scores and coverage depth, in a genome.
bigBed: A format similar to BED, but bigBed files are binary, compressed,
and indexed. Display of bigBed files in a genome browser is significantly
faster due to the compression and indexing, which allow transmittal of
only the part of the file that is needed for the current view instead of the
entire file.
bigWig: A format for visualization of dense, continuous data, such as GC
content, in a genome browser. A newer format from the WIG format (see
below), bigWig is a compressed and indexed binary file format and loads
significantly faster.

383
384 Appendix I

CRAM: Standing for Compressed Reference-​oriented Alignment Map, CRAM


is an alternative reads alignment file format to SAM/​BAM. Designed by
the European Bioinformatics Institute, CRAM uses a reference-​based
compression scheme, that is, only bases that are different from the ref-
erence sequence are stored. As a result, CRAM files are smaller than
equivalent BAM files.
FAST5: Stores raw electrical signals recorded from an Oxford Nanopore
sequencer (such as MinION) for sequencing information extraction. Based
on the HDF5 file format (see HDF5) with an ONT-​specific schema. Besides
raw detection signals, FAST5 files can also contain basecalls in FASTQ
format after analysis and other information such as signal correction.
Unlike FASTA (see FASTA) or FASTQ (see FASTQ) files, FAST5 files are
binary and cannot be directly opened with a text editor. FAST5 is expected
to be replaced by a new file format, POD5, for improved write perform-
ance, decreased file size, and more streamlined downstream data analysis.
FASTA: A text-​based format for storing sequences. A sequence stored in the
FASTA format contains only two elements: a single-​line description (or
defline) and the sequence text. The defline starts with the “>” symbol,
followed by a sequence identifier and then a short description. The
sequence text is usually divided into multiple lines with each less than
80 characters in length. This format has its origin in the FASTA program
package developed in the late 1980s. Multiple sequences can be stored in
a FASTA file. FASTA files often have file extensions of .fa, .fasta, or .fsa.
FASTQ: The current de facto standard for storing sequencing data generated
from various NGS systems. It is a compact text-​based format containing
nucleotide base sequences and their call quality scores. Each read sequence
in a FASTQ file is represented by four lines of information. The first line
starts with the symbol “@,” followed by sequence ID and descriptor. The
second line is the read sequence. Line 3 starts with the “+​” symbol, which
may be followed by the sequence ID and description (optional). Line 4
lists basecall quality scores for each base in the read sequence. This format
was originally developed by the Sanger Institute. FASTQ files have file
extensions of .fq or .fastq. Compressed FASTQ files also have the suffix .gz
or .gzip from the compression utility used to create them.
GFF: General (or Generic) Feature Format. GFF is a tab-​delimited text file
format that describes how genes or other genomic features are displayed
in a genome browser. There are different versions of this format, with
GFF3 being the current version and GFF2 now deprecated. The GFF
format can be converted to the BED format (see BED).
GTF: Gene Transfer Format. A refined GFF format. Identical to GFF2.
GVF: Stands for Genome Variation Format. Used to describe sequence vari-
ation information. An alternative to VCF (see VCF). GVF is based on the
GFF3 format (see GFF) with additional pragmas and attributes.
Appendix I 385

HDF5 (or H5): Standing for Hierarchical Data Format version 5. HDF5 is an
open-​source file format designed to store and organize large and com-
plex data. The hierarchical structure it uses is similar to a file system, in
that its two major objects, groups and datasets, are similar to directories
and files, respectively. The FAST5 file format (see FAST5) used by Oxford
Nanopore sequencers and the single-cell RNA-​seq gene-​cell matrix data
used by the 10× Genomics Cell Ranger software are based on the HDF5
format.
MEX: Market Exchange format. In 10× Genomics single-cell RNA-​seq, the
MEX file format is used by the Cell Ranger software to output gene-​cell
data matrix (besides HDF5). This is a sparse matrix format because of the
large number of 0’s contained in the file. This file format comprises three
files, i.e., matrix.mtx that contains the gene-​cell barcode matrix, barcodes.
tsv for storing cell barcodes, and genes.tsv for genes.
PED: A file format used by PLINK (a toolset for genome-​wide association
analysis) that contains pedigree/​phenotype data.
SAM: Standing for Sequence Alignment/​Map, SAM is a standard NGS
reads alignment file format, describing how reads are mapped to a ref-
erence genome. It is a tab-​delimited text format and human-​readable.
SAM files can be converted into its compressed binary version (BAM)
for faster parsing and file size reduction. SAM files have a file extension
of .sam. An indexed SAM file also has an accompanying index file that
has an file extension of .sai.
SFF (Standard Flowgram Format): A type of binary sequencing file generated
by 454 sequencers. Can be converted to the FASTQ format using utilities
such as sff2fastq.
VCF: Stands for Variant Call Format. A commonly used file format for
storing variant calls. It is a tab-​delimited, human-​readable text format
that contains meta-​information lines, a header line, and data lines that
describes each variant.
WIG: Wiggle Track Format. It is used for displaying continuous data track,
such as GC content, in a genome viewer such as the UCSC Genome
Browser. The WIG format is similar to the bedGraph format (see above),
but a major difference between the two is that data exported from a WIG
track is not as well preserved as that from a bedGraph track. The WIG
format can be converted to bigWig (see above) for improved performance.
387

Appendix II
Glossary

5-​Methylcytosine (5-​mC): The most frequently observed form of epigenetic


DNA modification. Produced by the addition of a methyl group to the
fifth carbon of cytosine. Cytosine methylation reduces gene transcription
and regulates chromatin remodeling.
Algorithm: A well-​defined procedure that comprises a set of instructions for
solving a recurrent problem.
Alignment: Similarity-​ based arrangement of sequences. In NGS data
analysis, sequence reads are usually aligned against a reference genome
to locate their genomic origins.
Allele: One particular variant form of a gene that has a number of alternative
sequence variants.
Annotation: The process of providing biologically relevant information to a
piece of DNA or RNA sequence. Also refers to the biological information
itself that is attached to a sequence.
ASCII: Standing for American Standard Code for Informational Interchange,
ASCII provides a standard for encoding characters. Since a computer only
deals with numbers, each human-​readable character has to be encoded
with a unique number in a computer. An ASCII code is the numerical
representation of a character in a computer. For example, in the ASCII
table, the character “A” is represented by the number 65.
Assembly: A computational process to reconstruct a longer sequence from
short sequences.
Barcode: Unique short artificial sequence(s) attached to DNA molecules in a
sequencing sample. The use of barcode sequence(s) enable identification
of different samples when they are sequenced together in a mixture (i.e.,
multiplex sequencing). Also see Multiplex Sequencing and Demultiplexing.
Basecall Quality Score: A score assigned to each basecall in a sequence read
to quantify the confidence level of making the call. In NGS it is defined in
the same way as the Phred quality score originally developed for Sanger
sequencing. Also see Phred Quality Score.
Bisulfite Conversion: A chemical process that leads to the differentiation
of methylated cytosines from unmethylated cytosines. The treatment
by bisulfite converts unmethylated cytosines in DNA to uracil, while
methylated cytosines are not affected by this process. Bisulfite conversion

387
388 Appendix II

coupled with NGS is a major means to study genome-​ wide DNA


methylation. Also see Whole-​Genome Bisulfite Sequencing.
Burrows–​Wheeler Transform (BWT): A method of permuting the characters
of one string into another string. In NGS data analysis, BWT enables fast
reference genome searching through providing efficient compression
and indexing.
cDNA: Complementary DNA. Refers to DNA that is reversely transcribed
from and therefore complementary to an mRNA species.
CDS (Coding DNA Sequence): The region of DNA that is translated into
protein.
Cell Ranger: A set of single-cell RNA-​ seq data analysis pipeline tools
developed by 10× Genomics, to carry out steps such as sequence
mapping, generation of gene-​cell matrix, and downstream analyses such
as cell clustering.
ChIP-​Seq: Chromatin immunoprecipitation coupled with sequencing.
A major application of NGS for studying genome binding of DNA-​
interacting proteins such as transcription factors.
Codon: A tri-​nucleotide sequence of DNA or RNA that codes for a specific
amino acid or the signal for protein synthesis termination. There are a
total of 64 codons, with 61 specifying amino acids and 3 as termination
signals.
Contig: A contiguous segment of RNA or DNA sequence resulted from
assembly of a set of overlapping sequence reads.
Copy Number Variation (CNV): One type of genomic variation caused
by changes in copy number of a DNA segment, usually as a result of
deletion or duplication. CNV is a subcategory of structural variation
and involves DNA segments that are usually larger than 50 bp. Also see
Structural Variation.
Coverage: The average number of times that nucleotides in different genomic
positions appear in a sequencing dataset. Also known as Sequencing
Depth or simply Depth.
Demultiplexing: The identification and separation of sequencing reads
that are generated from different samples, based on the unique barcode
sequence(s) they carry, after a multiplex sequencing run. Also see Barcode
and Multiplex Sequencing.
Depth: Same as Coverage.
DNA Polymerase: A class of enzyme that catalyzes the synthesis of new
DNA strand from free nucleotides, using an existing DNA strand as
template. Many molecular techniques, including PCR and sequencing-​
by-​synthesis, are based on the use of DNA polymerases.
DNase: An enzyme that catalyzes the hydrolysis of DNA into oligonucleotides
or nucleotides.
Epigenome: Refers to chemical modifications to DNA and histones, which
provides additional regulation to genomic activity.
Appendix II 389

Exon: A stretch of nucleotide sequence that is part of a gene providing coding


information for protein synthesis. Exons are transcribed to and usually
retained in mRNA.
Exome: The complete set of exons in an organism’s genome.
False Discovery Rate (FDR): A measure of statistical significance after
correcting for multiple testing. It estimates the proportion of false
discoveries in the final list of findings. Among the various approaches
for multiple testing correction, FDR estimation offers a balance between
statistical stringency and rate of type II errors and therefore is widely
used for high-​ throughput genomics data analysis. Also see Multiple
Testing Correction.
GC Content: The percentage of guanines plus cytosines in a DNA/​RNA
sequence or genome.
Gene Expression: The process by which the information encoded in a gene’s
nucleotide sequence is used to direct the synthesis of a functional gene
product. The level of gene expression in a cell or population of cells is
represented by the abundance of its product. The composition of the
large number of gene products and their expression levels in a cell or
population of cells constitute gene expression profile of the host cell(s).
Genome: The complete set of DNA sequence in a cell or an organism. Contains
the complement of information needed to form and maintain the cell or
organism. Including both protein-​coding and non-​coding sequences.
Genotype Posterior Probability: The probability of a genotype given an
observed dataset, calculated from NGS reads and often with the use of
prior genotype information.
Gene Ontology (GO): An initiative to provide consistent description of gene
products using standardized vocabulary. Each gene product is described
by three structured ontologies that encompass their associated biological
processes, cellular components, and molecular functions.
Hidden Markov Model (HMM): Named after the Russian mathematician
Andrei Markov (1856–​1922), HMM is a commonly used machine learning
and data mining approach for signal processing and pattern recognition.
A Markov model is a statistical model that deals with observed
sequences and state transitions. In bioinformatics, HMM is often used
for basecalling, sequence alignment, and gene prediction.
High-​Performance Computing (HPC): A computer system that has the
capability to perform over one teraflop (1012) floating-​point operations
per second by the use of parallel processing.
Indel: A generic term for either insertion or deletion of nucleotide(s) in a
DNA sequence. Such insertion/​deletion events lead to DNA mutation
and sequence length change.
Indexing: The process of creating a data structure for fast search. Techniques
of indexing for sequence alignment include hashing (storing information
on where a particular subsequence can be found in a reference genome or
390 Appendix II

a large collection of reads), suffix array (that consists of lexicographically


sorted genomic DNA sequence suffixes), and BWT (permutation of a
genome based on suffix array).
Irreproducible Discovery Rate (IDR): A measure of experimental
reproducibility. Developed to evaluate the reproducibility between
replicates of a ChIP-​seq experiment, it calculates the rate of irreproducible
discoveries, i.e., peaks that are called in one replicate but not in another.
K-​mer: In genome assembly or sequence alignment, k-​mer refers to all the
possible subsequences of length k in a sequence read.
Library: Collection of many different DNA (or RNA) fragments that are
systematically modified for target DNA screening or high-​throughput
analysis (including NGS). Specifically, a sequencing library is a pool of
DNA (or RNA) fragments with universal adapters attached to their ends.
To construct a sequencing library, DNA (or RNA) molecules extracted
from a population of cells are usually randomly fragmented, followed
by addition of universal adapters to the two ends of the fragments.
Sequences in the adapters enable subsequent enrichment and high-​
throughput sequencing of the fragments.
Long Non-​Coding RNA (lncRNA): Non-​protein coding RNA species that
are over 200 nucleotides in length. In comparison to small RNAs.
Machine Learning: A branch of Computer Science that focuses on developing
software algorithms that provide computers the capability to learn and
make predictions on new data. Machine learning is built on computational
model construction from existing input data, which is then applied to
new data for generating predictions or decisions.
Mapping: The process of searching the sequence of a read against the
reference genome sequence to locate its origin in the genome. Also see
Alignment.
Mapping Quality: An estimation of the probability of mis-​aligning a read to
a reference genome. It is reported as a Phred-​scale quality score. Also see
Phred Quality Score.
Mate-​Pair Reads: Reads generated from two ends of a long DNA fragment.
To achieve sequencing of the two ends, the long DNA fragment is first
circularized and then fragmented. Paired-​end sequencing of the fragment
that contains the junction of the two ends generates mate-​pair reads.
MeDIP: Methylated DNA immunoprecipitation with anti-​5-​methylcytosine
antibody.
Metagenome: The collection of all the genomes contained in a microbial
community that consists of many individual organisms.
Metagenomics: Studies of all the genomes existing in a microbial community
as a whole without the need to capture or amplify individual genomes.
Also referred to as environmental or community genomics.
Microarray: A high-​throughput genomics technology based on the use of
predesigned detection probes that are printed or synthesized on a solid
surface, such as glass or a silicon chip, in a high-​density array format.
Appendix II 391

Minor Allele Frequency (MAF): Frequency of the least abundant allelic


variant in a population.
miRNA: MicroRNA. See Small RNA.
mRNA: Messenger RNA, which carries protein-​coding information in DNA
for protein translation. It acts as the intermediate between DNA and
protein. An important component of a transcriptome.
Multiple Testing Correction: Adjustment of statistical confidence based
on the number of tests performed. Multiple testing without such an
adjustment leads to high levels of false positives. For example, at a p-​
value of 0.05, performing 100 comparisons simultaneously will generate
5 positive outcomes simply by chance if a correction is not applied.
Commonly applied multiple testing correction approaches including
the Bonferroni adjustment (conservative) and False Discovery Rate
estimation. Also see False Discovery Rate.
Multiplex Sequencing: Simultaneous sequencing of multiple samples
together. The use of artificial barcode sequence(s) enables sample
identification. Also see Barcode and Demultiplexing.
Multireads: Reads that map to multiple genomic locations.
N50: The weighted mean contig size of a genome assembly. To calculate N50,
all contigs are first ranked based on their lengths, which is then followed
by adding the ranked lengths from the top downward. N50 refers to the
length of the contig that makes the total added length equal to or greater
than 50% of the assembly size. An often-​used metric of de novo genome
assembly quality.
NAS: Network Attached Storage. Specialized computer data storage server
providing data access to a variety of clients through network.
Non-Coding RNA: RNA species that carry out functions other than coding
for proteins. Examples include small RNAs and lncRNAs. Also see Small
RNA and Long Non-​Coding RNA.
Normalization: A mathematical procedure to correct for unwanted effects
of non-​intended factors and/​or technical bias (such as differences in
sequencing depth between samples in RNA-​seq). This procedure puts
focus on the biological difference of interest, and makes samples in
different conditions comparable.
Normalized Strand Correlation (NSC): A measure of signal-​ to-​
noise
ratio in ChIP-​seq. It is calculated as the normalized ratio between the
maximum strand cross-​correlation (at the fragment-​length peak) and the
background cross-​correlation. Also see Relative Strand Correlation (RSC).
Open Reading Frame (ORF): A continuous segment of DNA containing
nucleotide triplet codons that starts with the start codon (ATG) and ends
with one of the stop codons (TAA, TAG, or TGA).
Operational Taxonomic Unit (OTU): A common microbial diversity unit
used in metagenomics that may represent a species or a group of species.
OTUs are clustered together based on DNA sequence information alone.
392 Appendix II

Paired-​End Reads: Reads obtained from the two ends of a DNA fragment.
Since the length of the DNA fragment, i.e., the distance between the
reads, is known, use of paired-​end reads provides additional positional
information in mapping or assembly of the reads. In comparison to
Single-​End Reads.
Pathway: A succession of molecular events that leads to a cellular response
or product. Each of such events is usually carried out by a gene
product. Many biological pathways are involved in metabolism, signal
transduction, and gene expression regulation.
PCA: Principal Component Analysis. A dimensionality reduction technique
to help summarize and visualize large and complex datasets. PCA is
widely used in next-​gen sequencing applications such as bulk and single-
cell RNA-​seq.
PCR Bottleneck Coefficient (PBC): An index of sequencing library
complexity. It is calculated after the read mapping step as the ratio
between the number of genome locations to which only one unique
sequence read maps and the total number of genome locations to which
one or more unique reads maps. PBC measures the distribution of read
counts towards one read per location.
Phred Quality Score (Q Score): An integer value that is used to estimate
the probability of making an error, i.e., calling a base incorrectly. It is
calculated as Q =​−​10xlog(10)P(Err). For example, a Q score of 20 (Q20)
means a 1/​100 chance of making a wrong call. Q30 represents a 1/​
1000 chance of making a wrong call, which is considered to be a high-​
confidence score. Q scores are often represented as ASCII characters for
brevity.
Picard: A set of tools written in Java for handling NGS data and file formats.
Pileup: A file format created with SAMtools showing how each genomic
coordinate is covered by reference sequence-​matching or -​unmatching
bases from all aligned reads.
piRNA: Piwi-​interacting RNA. See Small RNA.
Polymerase Chain Reaction (PCR): A molecular biology technique that
amplifies the amount of a DNA or RNA fragment, with the use of specific
oligonucleotide primers that flank the two ends of the target fragment.
Promoter: DNA sequence upstream of the open reading frame of a gene. The
promoter region is recognized by RNA polymerase during initiation of
transcription. Contains highly conserved sequence motifs.
Proteome: The complete set of proteins in a cell, tissue, or organ at a certain
point of time. Proteomics analyzes a proteome via identifying individual
component proteins in the repertoire and their abundance.
Quality Score: See Basecall Quality Score.
Read: Sequence readout of a DNA (or RNA) fragment.
RNA-​Seq: Stands for RNA sequencing. Also referred to as whole
transcriptome shotgun sequencing. RNA-​seq is a major technology for
transcriptome analysis and a major application of NGS.
Appendix II 393

RNAi: RNA interference, i.e., inhibition of gene expression. RNAi is usually


mediated by small RNAs, which lead to degradation of specific mRNA
targets.
RNase: An enzyme that catalyzes the degradation of RNA molecules.
Reduced Representation of Bisulphite Sequencing (RRBS): An NGS-​
based experimental approach that determines DNA methylation
pattern in a reduced genome (usually to save costs). The reduced
representation of the genome is usually achieved by the use of
restriction enzymes.
rRNA: Ribosomal RNA, i.e., RNA species that are essential components of
the ribosome. They play key roles in protein synthesis. By quantity, they
are the most abundant RNA species in a cell.
Relative Strand Correlation (RSC): A metric of signal-​to-​noise ratio in ChIP-​
seq. RSC is the ratio between background-​ adjusted cross-​ correlation
coefficient at the fragment-​length peak and that at the read-​length peak.
Also see Normalized Strand Correlation (NSC).
RNA Velocity: A single-cell RNA-​ seq analysis approach for making
predictions on future cellular states in terms of the speed and direction
of their movement along a trajectory. This approach is based on detection
and comparison of unspliced pre-​mature transcripts that still contain
introns and spliced mature transcripts.
SAN: Storage Area Network. A type of Local Area Network (LAN) designed
to handle large data transfers.
Sanger Sequencing: The first widely adopted DNA sequencing technology.
Devised by Dr. Fred Sanger, it is based on the principle of sequencing-​by-​
synthesis with the use of dideoxynucleotides that irreversibly terminate
new DNA strand synthesis once incorporated. With the advent of NGS
technologies, this sequencing method has become the synonym of first-​
generation sequencing.
Scaffold: Ordered arrangement of de novo assembled contigs. The
relative positional relationships between contigs are inferred by
mate-pair, paired-end, or long reads. In a scaffold, while the order of
contigs is known, sequence gaps still exist between contigs.
Sequencing Depth: See Coverage.
Sequencing Library: See Library.
Seurat: An R toolkit for comprehensive single-cell RNA-​seq data analysis.
Single-​End Read: Sequence read generated from one end of a DNA fragment.
This is in comparison with paired reads generated from both ends of a
DNA fragment. Also see Paired-​End Reads.
Single Nucleotide Polymorphism (SNP): DNA sequence polymorphism due
to variation at a single nucleotide position. Different from the term Single
Nucleotide Variation (SNV), SNP only refers to SNV that is relatively
common in a population with frequency reaching a certain threshold
(usually 1%). Also see Single Nucleotide Variation.
394 Appendix II

Single Nucleotide Variation (SNV): DNA sequence variation that involves


change at a single nucleotide position, e.g., the sequence change from
ATTGCA to ATCGCA.
siRNA: Small interfering RNA. See Small RNA.
Small RNA: Also called small non-​coding RNA. The major categories of
small RNA are miRNA, siRNA, and piRNA. In comparison to mRNA
molecules, these RNA molecules are much smaller in size. Small RNA
plays important regulatory roles in cells through mediating RNAi. Also
see RNAi.
Splicing: The process of removing introns from primary RNA transcripts and
joining of exons to form mature mRNAs. Splicing can be conducted in
more than one way for many genes, and alternative splicing can lead to
the production of different mRNA species from the same gene through
retaining different combinations of exons (or even introns sometimes).
SRA: Sequence Read Archive (also called Short Read Archive) maintained by
the National Center for Biotechnology Information (NCBI). SRA is one
of the major archives of NGS data generated worldwide. Other publicly
available NGS data archives include the European Nucleotide Archive
(ENA) maintained by the European Bioinformatics Institute (EBI).
Strand Cross-​Correlation: In ChIP-​ seq, there is a shift in base position
between reads generated from the forward and reverse strands of DNA.
Strand cross-​correlation is a measure of this shift, and calculated as the
Pearson correlation coefficient between the forward and reverse read
counts at each base position when the reads on the two strands are
shifted toward and away from each other at different base shift. Also see
Normalized Strand Correlation and Relative Strand Correlation.
Structural Variation (SV): Large-​scale genomic change that include large
indel, inversion, translocation, or copy number variation. Different from
SNPs or small indels, SVs involve DNA segments that are usually larger
than 50 bp. Also see Copy Number Variation.
Trajectory Inference (TI): Inferring transcriptomic change of an individual
cell over time based off of a snapshot of gene expression of a population
of cells at a certain point of time that represent a continuum of transitional
cellular states. TI is built on the premise that cells in the continuum share
many common genes and their gene expression displays gradual change.
Transcript: An RNA molecule transcribed from a segment of DNA.
Transcription Start Site (TSS): The nucleotide site in a segment of DNA from
which RNA transcription is initiated.
Transcriptome: The complete set of RNA transcripts in a cell, tissue, or organ
at a certain point of time.
Transcriptomics: Studies of the composition of a transcriptome. Encompasses
identification of the large number of RNA species in a transcriptome
and determination of their abundance levels. Major transcriptomics
technologies include microarray and RNA-​seq.
Appendix II 395

Translation: The process of protein synthesis from mRNA. Carried out by


ribosomes.
tRNA: Transfer RNA. The function of tRNAs is to transfer amino acids to
ribosomes for protein synthesis according to the triplet genetic code.
t-​SNE: t-​ distributed Stochastic Neighbor Embedding. A non-​ linear
dimensionality reduction method. The t-​SNE process maps the cells to
a dimension-​reduced space that best preserves cell-​cell similarity in the
original data.
UMAP: Uniform Manifold Approximation and Projection. Another non-​
linear dimensionality reduction method. It has a similar procedure to
t-​SNE, but it differs in how cell-​cell similarity is computed to provide
better preservation of global cell-​cell relationships.
UMI: Unique Molecular Identifier. UMIs are essentially a large number
of randomly synthesized, unique nucleotide combinations that are
attached to DNA fragments before any PCR amplification steps during
sequencing library preparation. The purpose of using UMIs is to identify
PCR duplicated molecules for removal.
UTR: Untranslated region of an mRNA molecule. Can be located on either
the 5’ end, or the 3’ end, of the mRNA molecule.
Variant Calling: Identification of sequence difference at specific positions
of an individual genome (or transcriptome) in comparison with a
reference genome. Each variant usually has a corresponding Phred-​scale
quality score.
Whole-​Genome Bisulphite Sequencing (WGBS): An application of NGS
that determines DNA methylation pattern across the entire genome
using bisulfite conversion. Also see Bisulfite Conversion.
397

Index

A Ascorbic acid, 29
AsmVar, 227
Ab initio splice junction detection, 123
Assay for Transposase-​Accessible
A-​Bruijn variant, 279
Chromatin, 372
ABySS, 271, 279, 282
Assembling contigs, into scaffold, 281
ACDtool, 132
Assembly Likelihood Evaluation (ALE),
Adapter ligation, 61, 73, 74, 75
348
AI-​based decision support tools, 256
Association for Molecular Pathology
Alignment methods, 92
(AMP), 248
Allele-​specific expression (ASE) analysis,
ATAC-​seq data, 300
186
AT-​overhang-​based adapter ligation
ALLPATHS-​LG, 275, 282, 285
process, 75
error correction module, 275
ATP synthesis
reference-​assisted assembly approach,
cytoplasmic membrane, 11
285
proton gradient, 12
Alu element, 29
AU-​rich element, 43
Alzheimer’s disease (AD), 11, 31
Automated Cell Type Identification
Amazon, 107
using Neural Networks (ACTINN),
Amazon Web Services (AWS)
176
Management Console, 108
Average silhouette width (ASW), 164
American Association for Cancer
Avidity chemistry, 367
Research (AACR), 246
American College of Medical Genetics
B
and Genomics (ACMG), 248
AMP rules, 255 Balrog, 351
pathogenicity/​benignity evidence, Barcode, 73
combination of, 255 BaseRecalibrator, 217
American Society of Clinical Oncology Batch effects, correction, 163
(ASCO), 254 BatchQC, 129
ANNOVAR tool, 245 Bayesian approach, 178, 352
Anti-​oxidant response element (ARE), 26 Bayesian mixed, 315
AnVIL, 109 Bayesian modeling, scDD employs, 180
APP gene, 31 BaySeq, 130
Application Programmer Interfaces BBTools suite, 347
(APIs), 112, 246 BCFtools, 218
Application-​specific integrated circuit Bcftools mpileup, 218
(ASIC), 67 “BED-​file” mode, 308
ApplyBQSR, 217 BedGraph
ARAGORN, 351 file format, 332
Argonaute processing, 206 track files, 309
Artificial intelligence, in variant BEDTools, 309
reporting, 256–​257 Benjamini–​Hochberg approach, 301, 306
Artificial neural networks (ANNs), 111, Binning algorithms, 350
168, 217 Biocontainers, 372

397
398 Index

Biofuel generation, 342 by genome instability, 32


Bioinformatic algorithms, 111 vs. normal cell comparison, 118
Bioinformatics tool development, 369 Canonical Correlation Analysis (CCA),
Bio-​polymer molecule, type of, 20 163
Bioremediation generation, 342 Canu, 277
Bismark, 329 Carbohydrates, 5, 7–​8, 10, 12
Bis-​SNP, 331 5-​Carboxylcytosine (5caC), 30
Bisulfite mapping, 326 CardioClassifier, 254
Bisulfite sequencing, 329 CardioVAI, 254
applications, 329 Carnelian, 353
steps of, 323 CarveMe, 353
BitBucket, 262 Catalogue Of Somatic Mutations In
BLAST, seed-​and-​extend approach, 87 Cancer (COSMIC), 246
BMTagger, 347 CD79a gene, 174
Bonferroni correction, 132 CD79b gene, 174
Boolean models, 188 cDNA sequences, 284
Bowtie algorithm, 122, 326 Cech complex, 168
Bowtie2/​BWA, 298 Cell cycle disturbance, 32
genome mapping, 208 Cell identity annotation tools, 175
performs mapping, 89 Cell membrane, 5, 7
Bracken, 352 component, 5
BRAIN Initiative Cell Census Network general structure, 6
(BICCN), 174 proteins, 9
Branch expression analysis modeling Cell Ontology (CO), 176
(BEAM), 184 Cell Ranger, 156, 173
BRCA1 gene, 32 Cellranger count, 157
BRCA2 gene, 20, 32 Cell Ranger software, 154
BreakDancer, 226 Cell-​splicing junction, 187
Bridge amplification, 61 Cellular heterogeneity, 76, 145, 147, 162,
Bridger, de novo transcriptome 176, 185, 187, 239
assemblers, 125 Cellular system
Bruijn graph, 277, 278, 280 cells molecules, 4–​5
BSMAP masks, 328 challenges, 3–​4
BSmooth, 332 code of life, 3
BS-​Seeker2/​BS-​Seeker3, 329 intracellular structure, see Intracellular
BugSeq, 352 structures/​spaces
Burrows–​Wheeler transform (BWT), 89, perpetuation/​evolution of, 4
90 protein-​coding/​non-​coding
BUSCO, 284, 348 sequences, 14
BWA algorithm, 91, 370 research & development, 14–​15
systems biology, 13–​14
Cellular trajectory
C
inference of, 184
Ca2+​-​mediated cell signaling, 9 topologies, 182
CAMI II challenge, 351 Cellular transcription, landscape
Cancer of, 51
Cancer Variants interpretation, open Cellular transcriptome, deep
source (CancerVar), 256 sequencing, 50
Index 399

Centers for Medicare & Medicaid Chromatin immunoprecipitation


Services, 237 (ChIP), 77
Central dogma, 20 Chromatin remodeling, 25, 50
Centrifuge, 352 Chromosomal breakage, 32
CentriMo, 315 Circular consensus sequencing (CCS), 65
CHANCE, 298 Circular RNAs (circRNAs), 50
CheckM, 348 Circulating cell-​free DNA (cfDNA), 239
Chemical/​enzymatic conversion-​based Circulating tumor cells (CTCs), 239
DNA methylation sequencing, 328 Circulating tumor DNA (ctDNA), 239
Chemistry Batch Correction algorithm, cis-​Regulatory modules, 39–​40
164 cisTopic, 373
ChIPDiff, 312 Cistrome, 314
ChIP DNA, for library prep, 295 CLC Genomics, 96
ChIPpeakAnno, 313, 315 Workbench, 96, 109, 371
ChIPQC reports, 307, 308 Cleavage Under Targets and Release
ChIP-​seq data, 298, 308, 315, 321 Using Nuclease (CUT&RUN), 296
analysis, 26, 297, 300, 314 Clinical Interpretations of Variants in
appropriate control, 295 Cancer (CIViC), 256
background noise/​signal profiles, 299 Clinical Laboratory Improvement
basic steps of, 294 Amendments (CLIA), 237
data quality, 301, 309 Clinical sequencing testing, 258
differential binding analysis, 312 ClinVar, 246, 247
experiment, 296 Cloud computing
gene expression data, 313 for NGS data analysis, 108
library complexity, 297 virtualization technology, 107
locally weighted regression (LOESS) CNN-​Peaks, 307, 376
normalization approach, 311 CNVnator, 228
NGS-​based approaches, 40 Coded protein, 254
NGS-​based technology, 293 Coding sequences (CDS), 22
normalization methods, 311 College of American Pathologists (CAP),
NRF2-​regulated genes, 314 254
peak calling algorithms, 306 bioinformatics pipeline validation, 261
peaking calling, 370 precision/​reproducibility of, 261
“phantom” peak, 303–​304 ComBat, 129, 162
process, 293 ComBat-​seq, 130
QC tools, 298 Combinatorial probe-​anchor ligation
quantitative analysis, 311 (cPAL), 366
substeps of calling peaks, 305 Combinatorial probe-​anchor synthesis
workflow, 297 (cPAS), 366
ChIP-​seq reads Common Workflow Language (CWL),
distribution of, 302 372
mapping, 298 Comparative genomic hybridization
ChIP-​seq signal, 301 arrays (aCGH), 259
adjustment, 295 Complementary DNA (cDNA), 72
changes, 312 Complete Genomics, 366
distribution, 309 Computing, for NGS data analysis
Chlorophyll, 12 big data, 103
Chloroplast, 5 bioinformatics skills, 111–​112
400 Index

cloud computing, 106–​108 NGS technologies, see Early-​stage


computing power, 105–​106 NGS technologies
data sharing, 103–​105 RNA-​Seq, see RNA-​Seq
data storage, 103–​105 Data generation, clinical sequencing
data transfer, 103–​105 artificial intelligence, in variant
parallel computing, 110–​111 reporting, 256–​257
software, 108–​110 bioinformatics pipeline validation,
Continuous long-​read (CLR) mode, 65, 261–​263
66 clinical phenotype match, 246–​247
Convolutional neural network (CNN), expert review, 257
82, 219, 375 functional consequence, 245–​246
CoolMPS chemistry, 366, 367 germline variants, classification of,
Copy number variation (CNV), 28 248–​254
Cortex, 227 human disease, relationship evidence,
Cost savings, 107 246
CRAM files, 95 library preparation/​sequencing
CRISPRDetect, 351 approaches, 240–​242
CRISPRFinder, 351 mode of inheritance, 247
Critical Assessment of Metagenome patient counseling/​periodic report
Interpretation (CAMI), 349 updates, 260–​261
Cromwell, 372 patient sample collection, 238–​240
CSS normalization, 355 patient’s electronic health record, 260
Cuffdiff 2, 130 prioritization, 247–​248
Cufflinks, 126 occurrence, frequency of, 245
Cumulative-​sum scaling (CSS), 354 read mapping, 243
CWL-​Airflow, 372 secondary findings, 260
Cytoplasm, 5, 7, 9, 42 somatic variants, classification of,
Cytoplasmic mRNA, 158 254–​256
Cytoscape, 136 testing report, generation of, 257–​259
Cytosine variant calling, 243
chemical structures of, 325 variant filtering, 243
demethylation products, 325 variant ranking, 247–​248
methylation, 78 variant validation, 259–​260
Cytoskeleton, 5 DBChip, 313
Cytosol, 7, 9 DBG2OLC, 282
dbSNP, 217
De Bruijn graph approach, 348
D
DECIPHER, 247
Darwinian evolution, 28 DeconSeq, 347
DAS tool, 349 DecontX, 159
Data accuracy, biases Dedupe tool, 347
factors, in sequencing, 75–​76 Deep learning (DL), 81, 374
in library construction, 74–​75 DeepMAsED, 348
Data analysis DeepMP, 376
computing, see Computing, for NGS DeepPVP, 246
data analysis DeepTools2, 298, 307
DNA methylation sequencing, 326 DeepVariant, 219
machine learning, 374–​376 DE gene identification, 180
Index 401

Degenerate oligonucleotide-​primed PCR Direct library preparation (DLP), 373


(DOP-​PCR), 373 Directly Attached Storage (DAS), 104
DE genes Dirichlet-​multinomial distribution, 159,
detection tools, 131–​132 178
hierarchical clustering of, 135 Disease-​causing variant, 245
identification of, 129 Disease Ontology (DO) hierarchy, 247
De novo genome assembly, 77, 271, 277, t-​Distributed Stochastic Neighbor
284 Embedding (t-​SNE), 167
assemblers, 370 Division of labor, 3
assembly quality evaluation, 282–​284 DMRFinder, 334
contig assembly algorithms, 277–​280 DNA-​binding motif, 314
error correction, 274–​277 pattern, 310
future development, 284–​285 transcription factor NRF2, 314
gap closure, 281–​282 DNA coding, 26, 257
genome characteristics, 274–​277 DNA contamination sequences, 347
genomic factors, 272 DNA duplication enzymes, 46
limitations, 284–​285 DNA extraction, 343
long/​short reads, 271 DNA fragmentation, 74
NGS applications, 271, 272 double-​stranded, 74
polishing, 280 ligation biases, 74
scaffolding, 281–​282 PCR biases, 75
sequence data preprocessing, 274–​277 DNA hypomethylation, 33
sequencing strategies, 272–​274 DNA intelligent analysis (DIANA), 210
workflow, 275 DNA-​interacting proteins, 26, 39, 293,
De novo metagenome assembly, 347 315, 321
Deparaffinization, 238–​239 binding of, 310
DESeq, 334 ChIP-​seq data analysis, 315
DESeq2, 130, 178, 310 DNA methylation data, 29–​30, 321, 331,
DESJ-​detection, 186, 187 332
DEXSeq, 136 analysis, 332
DIAMOND, 350 data interpretation, 335
Dicer processing, 206 quantification, 331
DiffBind, 313 visualization, 331, 333
Differential expression (DE) analysis DNA methylation sequencing, 370
methods, 178 bisulfite conversion, 322–​324
RNA-​seq, 310 ChIP-​seq peaking calling, 370
single-​cell tools, 179 cytosine methylation, from
Differentially Expression Genes demethylation products, 325–​326
Elimination Strategy (DEGES), 128 data analysis; see also DNA
Differentially methylated cytosines and methylation data
regions (DMCs/​DMRs), 332 data generation, 334
Differential network analysis, 189 data verification/​validation/​
Differential splicing (DS), 136 interpretation, 334–​335
DiffReps, 310, 312 demethylation products,
DiffSplice, 136, 137 quantification of, 330–​331
Diffusion maps, 167 differentially methylated cytosines
Diffusion Pseudotime (DPT), 182 and regions (DMCs/​DMRs),
Digital Cell Sorter, 174 332–​334
402 Index

enrichment-​based methyl-​seq, 324–​325 DNA target, 18


enzymatic conversion methyl-​seq, 324 DNA template, strands of, 19, 37
epigenomics, 321 DNBSEQ, 366
preprocessing, 326 DNBSEQ-​G50, 366
quality control (QC), 326 DNBSEQ-​G400, 366
read mapping, 326–​330 DNBSEQ-​T7, 366
reduced representation bisulfite DNB sequencing, error profile, 366
sequencing (RRBS), 323–​324 DoubletFinder, 158
strategies, 321–​322 DrImpute, 165
visualization, 331–​332 Drop-​seq, 146
whole-​genome bisulfite sequencing Drosha, 47
(WGBS), 322–​323 Duplex Sequencing, 242
DNA methylation study (Methyl-​Seq), Duplex-​specific nuclease (DSN), 120
77–​78 Dynamic Read Analysis for GENomics
DNA methyl-​seq approaches, 322 (DRAGEN) platform, 243
DNA methyltransferases, 30 Dysfunction, 32
DNA mutations, 27
DNAnexus, 371
E
DNA nucleotide sequence, 18
DNA packaging/​unpackaging, 25, 26 Early-​stage NGS technologies, 81–​98
DNA polymerases, 18, 39, 58–​59, 70, 321 algorithms/​reference genome
DNA probes, 117 sequences, 91–​93
DNA-​protein interactions, 26 approaches, 86–​91
DNA recombinase A, 351 basecalling, 81–​84
DNA recombination, 29 base quality score, 81–​84
DNA repair enzymes, 26, 46 data quality check, 81
DNA repair genes, 32 data quality control/​preprocessing,
DNA replication process, 19, 21, 27 84–​86
cellular system, perpetuation/​ FASTQ file format, 81–​84
evolution of, 4 file examination/​operation, 95–​97
fidelity of, 18–​20 Q-​scores, 84–​85
DNA/​RNA fragments, 68 read length distribution:, 85
DNA/​RNA polymerases, 26 read mapping, 86
DNA’s double helix structure, 18 SAM/​BAM, as standard mapping file
DNase I hypersensitivity sites, 229 format, 93–​95
DNA sequence, 57–​60 sequencing library, 85
access, 25–​26 tertiary analysis, 98
base sequence, 17 user-​friendly interface, 85
complexity, 342 EBSeq, 130
data, 221 E. coli, 23, 38
double helix structure, 17, 18 EdgeR, 130, 334
mutation, 27–​28 EggNOG-​mapper, 352, 353
replication process, 18–​20 Elastic Compute Cloud (EC2), 108
DNase-​seq data, 300 Element Biosciences technologies, 367
DNAstar, 371 Empty droplets vs. those containing
DNA storage, well-​protected cells, detection of, 160
environment, 6 EmptyDrops, 159
DNA strands, 17 Emulsion MDA (eMDA), 373
Index 403

ENCODE Consortium, 309 FASTA format, 280


ENCODE Software Tools, 298, 301 Fast Healthcare Interoperability
Encyclopedia of DNA Elements Resources (FHIR), 260
(ENCODE), 51 FastQC tool, 86
Endoplasmic reticulum (ER) FASTQ data, quality control (QC) of, 154
lumen, 9 FASTQ files, 64, 69, 154
organelles, 5 FASTQ format, 81–​83, 280
Endosome, intracellular structures/​ gigabytes (GB), 103
spaces, 8 terabytes (TB), 103
Endosymbiotic cyanobaterium, 12 Fastq-​join, 274
Enhancer RNAs (eRNAs), 50 FateID, 183
Enzymatic conversion, 326 Fatty acids, 5
bisulfite-​based chemical method, 324 FFPE-​compatible nucleic acid extraction,
DNA methyl-​seq data analysis, 324, 238–​239
327 Filtering variants, 249–​253
Enzymes, DNA repair, 18, 27 FindMarkers() function, 173, 180
EpiExplorer, 335 FindMotifs module (HOMER), 314
Epigenome, 29–​30 FindVariableGenes method, 166
Epigenomic studies, 14, 77–​78 Fisher’s exact test, 334
Error correction modules, 275 FLASH2, 274
Error-​prone sequencing, 272 Fluorescence-​activated cell sorting
Eukaryotic cells, 5, 10 (FACS), 148
structure of, 6 Fluorescence-​emitting core, 367
transcription, 39 Fluorescent dye, type of, 367
Eukaryotic genomes Fluorescent in situ hybridization, 238
gene expression, regulation of, 44 Food and Drug Administration (FDA),
non-​coding DNA elements, 21 237
Eukaryotic mRNA decay, 43 Formalin-​fixed paraffin-​embedded
Eukaryotic mRNA degradation, (FFPE), 119, 238
regulation of, 43 5-​Formylcytosine (5fC), 30
Eukaryotic ribosomes, 9 Fos genes, 150
Eukaryotic RNA polymerases, 39 FoundationOne CDx (F1CDx) panels,
Exome sequencing, 241 241
Exomiser, 246 FQC Dashboard tool, 86
Exons, 22, 23, 40 Fraction of reads in peaks (FRiP), 309
exon-​first methods, 123 FragGeneScan, 351
sequencing of, 23 FreeBayes, 185
Expectation-​maximization algorithm., 126 FRiP values, 309
eXpress, 126 Functional Annotation Of the
Expression recovery, 164 Mammalian (FANTOM) genome, 51
eXtasy, 246 Functional Genomics Data Society, 104
Extracellular signal, transduction of, 13 fusA (protein chain elongation factor),
351
F Fuzzy Bruijn graph (or FBG), 280
Factorial single-​cell latent variable
G
model (f-​scLVM), 167
False discovery rate (FDR), 132, 301 Galaxy system, 109, 372
Family wise error rate (FWER), 132 GATK BaseRecalibrator, 217
404 Index

GATK HaplotypeCaller/​BCFtools, 221 disease risk, 30–​33


GATK IndelRealigner, 217 epigenomic/​epigenetic diseases,
GATK Mutect2, 219 32–​33
GATK RealignerTargetCreator, 217 genome instability, 32
Gaussian distribution, 180 Mendelian (single-​gene) diseases, 31
Gaussian kernel, 167 Genome STRiP, 227
Gaussian mixture model (GMM), 349 Genomic DNA, to protein, 20–​21
G4 benchtop sequencer, 368 Genomic heterozygosity, 272
Gel beads-​in-​EMulsion (GEMs), 147 Genomic imprinting, 30
Genalice, 243 Genomic landscape
GenBank, 86 genome sizes, 21–​22
Gene calling, from metagenomic human genome, composition of, 24
sequences, 352 minimal genome, 21
Gene-​cell count matrix, 157 non-​coding genomic elements, 23–​24
Gene-​centered disease models, 32 protein-​coding regions, 22–​23
Gene coding, 22 Genomic polymorphisms, 14
Gene duplication, 28, 29 Genomic Regions Enrichment of
Gene fusion, 138 Annotations Tool (GREAT), 313, 335
GeneMANIA, 136 Genomics Evidence Neoplasia
GeneMarkS-​2, 351 Information Exchange (GENIE), 246
Gene Ontology (GO), 132, 174, 211, 248 Genomic variant cataloging projects, 215
Generalized linear model (GLM), 132, Genomiser, 246
161 GeoMx Digital Spatial Profiler from
Gene regulatory network (GRN), 187 Nanostring, 374
inference methods, 188 Germline cells, 27
scRNA-​seq data, 187, 188 Germline SNVs, 259
GeneReviews, 259 GFAP (glial fibrillary acidic protein)
Gene Set Enrichment Analysis (GSEA), gene, 174
136 GhostKOALA, 353
GeneStudio S5, 70, 72 Gibbs motif sampler, 314
Genetic circuit, 13 Gigabytes (GB)
Genetic mutation/​variation de novo assembly, 106
identification, 77 FASTQ format, 103
Genetic risk assessment, 371 Gilbert’s method, 57
Gene transcription, 21, 22, 126 GitHub, 262
Gene translation, 21 Glimmer-​MG, 351
Genexus, 70 Global Alliance for Genomics and
Genome assembly, error correction, 280 Health (GA4GH), 260
Genome evolution, 28–​29 Global positioning, 87
Genome in a Bottle (GIAB), 242 Glycolytic pathway, 10
Genome instability, 185 Golgi apparatus, 5, 8–​10
Genome modulation Google, 107
nucleotides/​histones, chemical Google Cloud Platform (GCP), 109
modifications, 77 Graphics processing unit (GPU), 106
regulatory mechanism, 77 Greedy approach, 277
GenomeScope, 276 Green pigment, 12
Genome sequencing GridION data, 68
Alzheimer’s disease (AD), 31–​32 GroopM, 349
Index 405

GSMMs, 353 Human genomic DNA, 25


GTDB-​tk, 352 Human/​interaction PHIVE (hiPHIVE),
GULO gene, 29 247
Guppy, 69 HUMAnN, 353
gVCF format, 223 Human Phenotype Ontology (HPO)
gyrB (DNA gyrase subunit B), 351–​352 project, 246
Huntington’s disease, 31
H Hybrid assemblers, 280
HybridSPAdes, 348
HaplotypeCaller, 185, 218, 221
Hybrid-​super-​transcriptomes, 158
Hardy–​Weinberg equilibrium, 223
Hydrogen peroxide, 8
Health Level Seven International (HL7),
260
I
Heart disease, 11
Helicase, 18 ICF (immunodeficiency, centromeric
Heligmosomoides polygyrus, 177 instability, and facial anomalies)
Hematoxylin & eosin staining, 238 syndrome, 32
Heteroplasmy, 11 IDBA-​Hybrid, 285
HiCanu, 279 Illumina
Hi-​C data, 282 DRAGEN reads mapper +​variant
Hidden Markov models (HMMs), 111, caller system, 374
313 Infinium MethylationEPIC
Highly variable genes (HVGs), 166 BeadChips, 321
High-​mobility group (HMG) proteins, Isaac Genome Alignment Software, 87
26 platforms, 69, 76
High-​molecular-​weight (HMW), 342 sequence data generation process, 63
High-​performance computing (HPC) sequencing flow cell, 61
systems, 105 sequencing technology, 60–​63
HISAT, 123–​4 small RNA sequencing protocol, 207
HISAT2, 123, 328 Illumina–​generated FASTQ files, 83
Histone H3 lysine 27 trimethylation, Illumina reversible terminator
295 short-​read sequencing
H3K27me3, 295 cost, 63
HOMER, findPeaks module, 306 data output, 63
Homopolymer error, 69 error rate, 63
HoneyBADGER, 186 Illumina sequencing technology, 60
HPC cluster, 112 implementation, 60–​63
HT-​Seq Python framework, 126 read length, 63
HTT gene, 31 sequence data generation, 63–​64
Human Cell Atlas, 165, 174 Immunohistochemistry, 238
Human Gene Mutation Database Immunoprecipitated DNA fragments,
(HGMD), 246 300
Human genome, de novo assembly, 274 Immunoprecipitation, 295
Human Genome Organization Gene Independent component analysis (ICA),
Nomenclature Committee (HGNC), 166
257 Ingenuity Pathway Analysis (IPA), 136
Human Genome Project, 14, 31 Integrated Microbial Genomes &
Human Genome Variation Society Microbiomes system (IMG/​M),
(HGVS), 257 352–​353
406 Index

Integrative Genomics Viewer (IGV), 97, K


259
Kaiju, 352
Integrative non-​negative matrix
KGGSeq, 247
factorization (iNMF), 164
K-​means clustering algorithm, 172
InterPro, 352
K-​mers, 87, 275
Intestinal epithelium, cellular
coverage profile of, 276
composition, 177
hash table-​based reference genome
Intracellular structures/​spaces
indexing, 87
cell membrane, 6–​7
seed-​and-​extend methods, 123
chloroplast, 12
KneadData, 347
cytoplasm, 7–​8
K-​nearest neighbor (kNN) method, 158
cytoskeleton, 10
Kraken, 352
endoplasmic reticulum, 9
Krebs cycle, 10, 11
endosome, 8
Kruskal–​Wallis rank-​sum test, 355
Golgi apparatus, 9–​10
Kruskal–​Wallis test, 332
lysosome, 8
mitochondrion, 10–​11
L
nucleus, 5–​6
peroxisome, 8 LanceOtron, 307
Intra-​experiment batch effects, 164 Lancet, 219
Introns, 40 LEfSe, 355
Ion 318 Dx chip, 72 Leiden methods, 172
Ion Torrent family, 70 Limit of detection (LOD), 240
Ion Torrent platform sequences, 63, 69 Linear dimensionality reduction
Ion Torrent semiconductor sequencing methods, 167
system, 69 Linked Inference of Genomic
cost, 70–​72 Experimental Relationships
date output, 70–​72 (LIGER), 164
error rate, 70–​72 Lipid bilayer structure, 6
implementation, 70 Lipids, 5
read length, 70–​72 lncRNAs, biogenesis of, 50
sequence data generation, 72 Locally Linear Embedding (LLE), 167
sequencing principle, 69–​70 Logistic Regression classification, 375
Irreproducible Discovery Rate (IDR), Long Fragment Read (LFR), 273, 369
296, 298, 308 Long-​non-​coding RNAs (lncRNAs), 45
experimental reproducibility, 296 LoopSeq, from Loop Genomics, 369
six peak callers, performance of, 308 Louvain community detection method,
Irritable bowel syndrome (IBS), 376 172
Isoform-​centric methods, 136 Louvain methods, 172, 182
isomiRs, 209 Lysosome, 8
intracellular structures/​spaces, 8
J lysosome membrane, 8
JASPAR, 315
M
Java-​based tool, 256
Jellyfish, 275 Machine learning (ML), 374–​376
JointSNVmix, 219 algorithms, 256
Jumping genes, 24 Mac OS, 109
Jun gene, 150 MACS2 shifts, 300–​301, 306
Index 407

“Major-​allele” reference genome, 93 Mendelian disorders, 238, 248


MAMBO, 353 MERFISH, from Vizgen, 374
Mann–​Whitney U test, 332 Messenger RNAs (mRNAs), 8
MAnorm2, 311, 313 binding proteins, 50
Mapping and Assembly with Qualities double-​stranded miRNA, 47
(MAQ), 87 miRNA-​mRNA interaction, 48
Mapping process, 96 precursor of, 47
MAPping program, 323 primary transcript (pri-​miRNA), 47
Mapping protein-​DNA interactions stability regulatory sequences, 43
ChIP-​Seq, principle of, 293–​295 suppressing target mRNA activity,
data quality control, 297–​300 48
differential binding analysis, 310–​313 trafficking of, 44
experimental control, 295 MetaBAT 2, 349
functional analysis, 313–​314 MetaBinner, 349
integrated ChIP-​Seq data analysis, 315 MetaGeneAnnotator, 351
library preparation, 295–​296 Metagenome-​assembled genomes
motif analysis, 314–​315 (MAGs), 345
peak calling, 300–​307 Metagenomes
peak visualization, 309 analyzer, 349
post-​peak calling quality control, characterization, 370
307–​309 metabolomic analysis of, 353
read mapping, 297–​300 steps of, 346
replication, 296 MetagenomeSeq, 355
sequencing length/​depth, 296 Metagenomics, 78, 341
Mapping quality score, 94 metagenomics RAST server
MarginPolish & HELEN, 280 (MG-​RAST), 352
Marker genes, expression of, 174 MetaMaps, 352
Marker profiling, 240 MetaPhlAn, 351, 352
Markov model, 315 MetaQUAST, 348
MARVEL, 277 MetaSPAdes, 348
MaSuRCA, 282, 348 MetaWatt, 349
Mate-​pair sequencing, 273 MetaWRAP, 349
MATLAB, 112 Methylated-​CpG Island Recovery Assay
Mature miRNA species, 206 (MIRA), 324
MaxBin 2.0, 349 Methylated cytosines/​regions, tools for
MaxEntScan, 245, 248 detection, 334
MBLASTX, 353 Methylation bias, 330
Mean-​variance relationship, 162 methyl-​CpG-​binding domain capture
MECAT, 277 (MBDCap), 324
Medaka, 280 5-​Methylcytosines (5mCs), 29, 321
MeDIP-​seq, 324, 331 MethylKit, 334
MedlinePlus Genetics, 259 MethylSig, 334
MEGANIZER, 350 Methy-​Pipe, 332
MelonnPan, 354 MGI/​BGI nanoball sequencing, 367
MEME-​ChIP, 315 MGnify, 353, 356
Memorial Sloan Kettering–​Integrated Microbial community profiling, 345
Mutation Profiling of Actionable Microsatellite instability (MSI), 241
Cancer Targets (MSK-​IMPACT), 241 Microsoft, 107
408 Index

MinHash alignment process (MHAP), NCBI


277 Prokaryotic Genome Annotation
Minimap2, 92 Pipeline (PGAP), 352
Minimum spanning tree (MST), 181 SRA database, 355
MinION data, 68 SRA repository, 105
MinKNOW software, 69 Needleman–​Wunsch algorithm, 91
Minor allele frequency (MAF), 245 Negative binomial (NB), 161
MinPath, 353 distribution, 147
MINSEQE (Minimum Information about Poisson, 301
a high-​throughput Nucleotide Neonatal intensive care unit (NICU), 237
SEQuencing Experiment), 105 Network Attached Storage (NAS), 104
miRanda, 210 Network speed, 104
miRDeep2, 210 NeuralPolish, 280
MiSeq, 76 NeuSomatic, 219, 220
Mitochondrial DNA (mtDNA), 11 NextDenovo, 279
Mitochondrion, 5 Nextera-​based method, 153
mnSBS chemistry, 368 Nextera XT protocol, 344
ModelSEED, 353 Nextflow, 372
Molecular barcoding, 242 Next-​generation sequencing (NGS)
Monocle 3, 182 technologies
Monogenic/​Mendelian diseases, 31 analytic workflow management,
Mouse Brain Atlas, 174 370–​372
Mouse Genome Informatics (MGI), 247 applications of de novo genome
Multi-​Dimensional Scaling (MDS), 167 assembly, 77
Multiple annealing and loop-​based biases/​factors, in sequencing, 75–​76
amplification cycles (MALBAC), 373 biases, in library construction, 74–​75
Multiple displacement amplification bioinformatics tools, evolution/​
(MDA), 373 growth of, 369–​370
Multiplex ligation-​dependent probe changing landscape, 365–​366
amplification (MLPA), 259 comparative analyses, 354
MultiQC tool, 86 cost, 63, 65, 68–​72
Multireads, 95 data analysis, see Computing, for NGS
MutationAssessor, 246 data analysis
MutationTaster, 246, 247, 256 date output, 68–​72
Mutect2, 220 de novo assemblers, 272
MutPred, 246 de novo genome assembly, 77
Mutual nearest neighbors (MNN), 163 data output, 63, 65
Mycoplasma genitalium, 21 DNA sequence, 57–​60
early-​stage, 81–​98; see also Early-​stage
NGS technologies
N
epigenomics/​DNA methylation study
NanoCaller, 219 (Methyl-​Seq), 77–​78
on deep CNN, 375 error rate, 63, 65, 68–​72
Nanopolish, 280 experiment, workflow of, 73
Nanopore sequencing, 67 files transferring, 104
Nanopore Technology, 243 general overview of, 82
NanoQC, 86 genetic mutation/​variation
Nasuia deltocephalinicola, 22 identification, 77
Index 409

Illumina sequencing technology, 60 OnClass, 176


implementation, 60–​65, 68, 70 Oncogenic somatic variants, 256
machine learning, 374–​376 Online Mendelian Inheritance in Man
mapping of, 87 (OMIM), 246, 247
metagenomics, 78 Open reading frame (ORF), 20
platforms, comparison of, 71 metagenomic sequences, 351
protein-​DNA interaction analysis protein translation, 20
(ChIP-​Seq), 77 Operational taxonomic units (OTUs),
read length, 63, 65, 68–​72 342
read mapping, 88, 89 Orphanet, 247
RNA sequencing, 36 Overlap-​Layout-​Consensus (OLC)
seed sequences, 90 approach, 277, 278
sequence data generation, 63–​66, 69, Oxford Nanopore sequencing, 227
72 Oxford Nanopore technologies (ONT)
sequencing principle, 64, 67–​70 cost, 68–​69
sequencing technologies, 366–​369 data output, 68–​69
single-​cell/​spatial sequencing, error rate, 68–​69
372–​374 implementation, 68
transcriptomic profiling, 76 nanopore sequencing, 345
trends, 365–​366 platforms, 91, 348
whole-​genome sequencing, 271 read length, 68–​69
workflow, 72–​74 sequence data generation, 69
NextPolish, 280 sequencer, 280
NHLBI Exome Sequencing Project (ESP), sequencing principle, 67–​68
245
NNSplice, 245 P
NOISeq, 132, 210
PacBio, 91, 227, 272, 348, 370
Non-​coding RNAs, 47, 50
PacBio data
genes, 23
CCS/​ONT, 274
species, 51
holistic kinetic (HK) model, 376
Non-​negative matrix factorization
PacBio HiFi, 279
(NMF), 167
PacBio platforms, 69, 95, 120, 219
Non-​redundant fraction (NRF), 298
PacBio sequencing library preparation/​
Normalized strand cross-​correlation
sequencing, 66
(NSC), 298
PacBio SMRT sequencers, 326
NovaSeq, 63
continuous long reads, 65
Novoalign, 87
nanopore sequencing, 345
Nucleic acids, 5
Paired-​End Mapper (PEMer), 226
Nucleobases
Paired-​end sequencing, 63
Rungs, 17
Pair-​Hidden Markov Model (PairHMM),
types of, 17
218
Nucleotide-​specific antibodies, 366
PANDAseq, 274
Paris japonica, 22
O
Parkinson’s disease, 11
Oases, de novo transcriptome assemblers, Partition-​based graph abstraction
125 (PAGA), 170
Oligo-​dT primers, 153 cellular trajectory topologies, 182
Omniome, 368 performing methods, 184
410 Index

trajectory inference methods, 183 Portability and Accountability Act


Partitioning Around Medoids (PAM), (HIPAA), 262
132 Potential of heat diffusion for
“Patching” module, 277 affinity-​based transition embedding
Pathogenic/​Likely Pathogenic variants, (PHATE), 170
260 PrecisionFDA Truth Challenge V2, 374
PCR bottleneck coefficient (PBC), 297 Precision Medicine Knowledge Base
PCR, sequencing-​by-​synthesis process, (PMKB), 256
75 Principal components analysis (PCA),
PeakSeq, 306, 307 129, 166
PEAR, 274 Prodigal, 351
Pearson correlation coefficient (PCC), Prokaryotic cells, 5, 6
296 DNA packaging, 25
PEPPER-​Margin-​DeepVariant pipeline, nucleoid of, 25
375 Prokaryotic genes, transcription, 37
Perl, 111 Prokaryotic ribosomes, 9, 45
Peroxisome, intracellular structures/​ Prokaryotic transcription, regulation of,
spaces, 8 38
PGM Dx system, 70 Prokka, 352
PhastCons, 256 PromethION data, 68
Phen-​Gen, 246 PROPER, 121
PhenIX, 246 Protein coding sequences, 28
Phenolyzer, 246 genes, 23, 39, 50, 126
Phenomizer, 246 gene sequences, 23
Phevor, 246 mRNA transcripts, 51
PhyloP, 248, 256 regions, 22, 243
Picard, 97 Protein-​DNA complex, 293
PierianDx, 371 affinity binding, 325
Pileup format file, generation of, 97 Protein-​DNA interaction analysis (ChIP-​
Pilon, 280 Seq), 43, 77
piRNA clusters, 49 Protein-​made skeletal structure, 10
piRNA precursor, 49 α-​Proteobacteria, 11
PIWI-​interacting RNAs (piRNAs), 45 Proteomics, 14
PIWI proteins, 49 PSEN1 gene, 31
Plastid, 12 PSEN2 gene, 31
Plastid DNA (ptDNA), 12 Pseudoalignment, 156
PlotCorrelation, 300 Punctate-​binding protein factors, 313
Point mutations, 27 PycoQC, 86
Poisson distribution, 130–​132, 148 Pyrosequencing, 259
Poisson/​negative binomial Python, 111
distributions, 301
Poisson p-​value thresholds, 306
Q
POLCA, 280
Poly-​A polymerase, 42 QIAGEN Clinical Insight (QCI), 254
Polymorphisms, 27–​28 interpret, 371
DNA base sequence, 17, 95 Qiagen’s QCI interpret, 256
genome-​wide sequence, 14 Qualimap 2, 97
PolyPhen, 246, 248, 256 Quality-​aware detection, 276
Index 411

Quality control (QC), 326 RNA interference (RNAi), 45


metrics, 297 RNA molecules
tools generation/​processing/​turnover, 36
library preparation, 242 DNA template, strands of, 37
post-​mapping data, 97 eukaryotic genes, pre-​mRNA
Quality score (Q-​score), 83–​85 transcription of, 38–​40
data quality, metrics of, 84–​85 localization, 42
encoding of, 84 mRNA, maturation of, 40–​42
low-​quality basecalls, 85 mRNA transcript level regulation,
read length distribution, 85 43–​44
sequencing library, 85 prokaryotic genes, transcription of,
QUAL value, 223 37–​38
Quantapore, Base4 sequencing, 368 stability/​decay, 42–​43
Quantitative Insight Into Microbial transport, 42
Ecology (QIIME2), 355 as messenger, 35, 44
long non-​coding RNAs, 50
miRNA, 47–​49
R
non-​coding RNAs, 50–​51
Racon, 280 piRNAs, 49–​50
RaGOO, 285 ribozyme, 45–​46
Random forest (RF), 176 RNAi/​small non-​coding RNAs, 47
Random Forest-​based classifier, 376 siRNA, 49
RAVEN, 353 snRNA/​snoRNA, 46
Ray Meta, 348 telomere replication, 46
RD-​based algorithms, 228 molecular structure of, 35–​36
Read mapping tools, 328 RNA polymerase, 37–​39
reads per million (RPM), 210 RNA polymerase II, 295
Real-​Time Analysis (RTA) software, 63 types of, 38
Recurrent neural networks (RNNs), 81 RNA-​seq, 117, 119, 127
Relative strand cross-​correlation algorithm-​related publications, single
coefficients (RSC), 298 cell, 371
Resequencing data, workflow for analysis tools, 128
genotyping/​variation discovery, capabilities, 120
216 differential expression, 310, 313, 334
Ribonuclease, 47 experimental design, 118
Ribonucleoprotein (RNP) complexes, 42 factorial design, 118
Ribosomal RNAs (rRNAs), 9, 120 gene-​level analysis, 76
Ribosomes, 9 principle of, 117
Ribozymes quantification of reads, 126
to protein enzymes, 45 randomization, 118–​119
spliceosome, 46 read mapping, 122–​126
RNA activation (RNAa), 205 replication, 118–​119
RNA-​binding proteins, 36 sample preparation, 119–​121
RNA enzymes, 36 sequencing library preparation, 119–​121
RNA expression analysis methods, 205 sequencing strategy, 121–​122
RNA-​induced silencing complex (RISC), total count approach, 354
47, 48 upper quartile approach, 354
RNA Integrity Number (RIN), 119 variant calling, 221
412 Index

RNA-​seq data, 129, 178, 284, 315 Sanger sequencing method, 57–​59, 77,
batch effect removal, 129 259, 271
data analysis, 98, 132 Savant, 223
data distribution, 121 Scaffolding algorithms, 281
DE analysis, 313 SCALE, 373
differentially expressed genes, Scanorama, 163
identification of, 129–​133 Scanpy, 173
differential splicing analysis, 136–​137 SCAN-​SNV, 373
discovery tool, 137–​138 scATAC-​seq data analysis tools, 373
experimental design, 118 SCcaller, 373
gene clustering, 134 SciDAP, 371
identified genes, functional analysis Scmap-​cell, 176
of, 134–​136 Scmap-​cluster mode, 176
multiple testing correction, 133–​134 scPred, 176
normalization, 127–​128 scRNA-​seq analysis, goal of, 171
overdispersion problem, 131 scRNA-​seq data analysis, 152, 176
reads, subsequent mapping of, 122 algorithms, 370
visualization of, 137 DE analysis, 179
RnaSeqSampleSize, 121 normalization approaches, 161
RNA-​seq sequencing libraries, 120 single nucleotide variation (SNV), 185
RNA-​seq study, 106 structure of, 154
rnaSPAdes, de novo transcriptome workflow, 155
assemblers, 125 scRNA-​seq tools, 181
RNA splicing, non-​canonical, 138 SeattleSeq, 229
RNA transcripts, 41, 117 Seed-​and-​extend methods, 86
“RNA world” hypothesis, 45 Self-​Organizing Map (SOM), 132
Roche’s NAVIFY Mutation Profiler, 256 Sentieon, 243
R package, 109–​111, 137, 307 SeqMonk, 97
rpoB (RNA polymerase beta subunit), 351 Sequence Read Archive (SRA), 104
rRNA depletion, 120, 125 Sequencing-​by-​desynthesis process, 368
rRNA genes, 351 Sequencing error correction, 274, 276
RSAT peak-​motifs, 314 Sequencing library preparation
RSEM, 126 protocols, 344
RTG Tools, 225 Seurat, 172, 173
Rungs, 17 Seurat Integration, 163
RUVSeq, 129 Seven Bridges, 371
Seven Bridges GRAF pipeline, 375
SGA, 282
S
Shannon entropy, in QDMR, 332
SAM/​BAM alignment section, 93 Shared Nearest Neighbor (SNN) graph,
SAM/​BAM files, 95, 97 172
file format, 93 Short Oligonucleotide Alignment
FLAG status, 94 Program (SOAP), 87
for storing NGS read alignment, 94 Short Tandem Repeats (STR), 240
SAMtools, 97, 185 Signac, 373
packages, 96 Signal imputation, 164
pileup file format, 97 Signal sparsity, challenges of, 186
Sanger sequence assemblers, 271 Signal-​to-​noise ratio, 368
Index 413

Signal transduction, 7 Single-​cell whole genome bisulfite


Silico variant filtering, 244 sequencing (scWGBS), 373
Single-​cell ATAC-​seq, 372 Single-​molecule real-​time (SMRT)
Single-​cell clustering methods, 173 sequencing, 59, 64
Single Cell Genotyper, 373 cDNA synthesis, 156
SingleCellNet, 176, 177 cost, 65
Single-​cell reduced representation data output, 65
bisulfite sequencing (scRRBS), 373 DNA samples, 64
Single-​cell RNA-​seq (scRNA-​seq), 145 error rate, 65
alignment/​transcript counting, 156–​157 implementation, 64–​65
alternative splicing analysis, 186–​187 PacBio sequencing, 64
batch effects correction, 162–​166 polyadenylation, 207
batch effects minimization, 149–​150 read length, 65
cell clustering, 171–​173 sequence data generation, 65–​66
cell identity annotation, 173–​177 sequencing principle, 64
cell number, 147–​149 SMRTbell, 65
compositional analysis, 177–​178 Single nucleotide polymorphism (SNP), 27
data clean-​up post alignment, 157–​160 Single nucleotide variation (SNV), 27
differential expression (DE) analysis, CaSpER, 186
178–​181 scRNA-​seq data, 185
dimension reduction, 166–​168 Single-​nucleus methylome sequencing
2-​D space, visualization of, 169 ver2 (snmC-​seq2), 373
experimental design, 146–​147 SingleR, 177
false-​negative detection, 145 Singular Genomics, 368
feature selection, 165–​166 SiPhy, 246, 256
general lab process, 151 Sliding linear model (SLIM) method, 334
gene regulatory network (GRN), Small interfering RNA (siRNAs), 45
187–​189 non-​protein-​coding RNAs, 45
high-​throughput approach, 146 suppressing target mRNA activity, 48
initial data preprocessing, 154–​156 Small nuclear RNAs (snRNAs), 40
lab process, 151 Small nucleolar RNAs (snoRNAs), 45
library construction/​sequencing, Small RNA sequencing, 205
153–​154 functional analysis, 210–​211
low-​throughput methods, 146 identification of, 210
normalization, 160–​162 mapping, 208–​209
overview of, 145–​146 normalization, 209–​210
preparation of, 150–​152 overview of, 205–​206
preprocessing of, 154 post-​transcriptional editing, 209
quality control, 154–​156 preprocessing, 207–​208
sample replication, 149–​150 putative identification, 209
sequencing depth, 147–​149 sequencing analysis, 206–​207
signal imputation, 164–​165 Smith–​Waterman algorithm, 91
single nuclei preparation, 152 SnapATAC, 373
SNV/​CNV detection/​allele-​specific snoRNAs, 46
expression analysis, 185–​186 SNP profile, 159
trajectory inference, 181–​184 SNVMix, 221
visualization, 168–​170 SOAPdenovo, 272, 347
workflow, 155 SOAPdenovo2, 282
414 Index

SOAPdenovo-​trans, de novo TargetScan, 210


transcriptome assemblers, 125 Taxon ID, 350
Somatic mutations Taxonomic profilers, 352
deep learning, 220 Taxonomy binning, 349
vs. germline variations, 220 Telomere, 24
SomaticSniper, 219 Telomere-​to-​Telomere (T2T) Consortium,
Somatic variants, 259 285
SoupX, 159 Tensorflow-​based framework, 375
SourceForge, 262 Terra, 372
SPAdes, 282, 369 TET-​assisted bisulfite sequencing
Spanning-​tree progression analysis (TAB-​seq), 325
of density-​normalized events Tet methylcytosine dioxygenase 2
(SPADE), 170 (TET2), 324
Spectral embedding, 167 TGS-​GapCloser, 282
Spike-​in controls, artificial, 128 de novo genome assembly, 282
SpliceAI, 245 workflow, gap closing, 283
16S rRNA amplicon sequencing, 343 ThermoFisher’s Ion Torrent
16S rRNA gene, 342 semiconductor sequencing, 59
16S rRNA PCR primers, 343 Thylakoids, 12
ssizeRNA, 121 Tissue-​of-​origin (TOO), 376
SSPACE-​LongRead, 281 Titin gene, 22
SSrGE, 373 TNscope pipelines, 375
Standardized standard deviation (SSD), Tomtom, 315
of genomic coverage, 309 TopHat/​TopHat2, exon-​first methods,
Standard Query Language (SQL), 112 123
Standard univariate analysis methods, 178 Total-​sum scaling (TSS), 354
Stochastic effects, 373 Trans-​ABySS, de novo transcriptome
Storage Area Network (SAN), 104 assemblers, 125
Strelka2, 185, 219, 220 Transcriptional regulation, 50
StringApp, 136 Transcription start site (TSS), 37
StringTie/​StringTie2, de novo Transcriptome, 23, 152
transcriptome assemblers, 125 Transcriptomics
Structural variant (SV) calling, 27 gene/​transcript expression, 126
calling, steps of, 226 profile, of biological sample, 76
CNV detection algorithms, 227–​228 by RNA-​Seq, see RNA-​Seq
detection algorithms, 226, 227 single-​cell RNA-​Seq, see Single-​cell
integrated analysis, 228 RNA-​Seq
long reads, 227 Transfer RNAs (tRNAs), 11
short reads, 225–​227 Transitions and transversions (Ti/​Tv)
SR/​AS approaches, 227 ratio, 224
Support vector machine (SVM), 111, 176 Transposable elements, 24
Supra-​macromolecular machines, 8 Transposase Enzyme-​Linked Long-​read
SVDetect, 227 Sequencing (TELL-​seq), 273, 369
Trim Galore, 326
Trinity CTAT, 185–​186
T
tRNAscan-​SE, 351
TABLE_​ ANNOVAR script, 229 t-​SNE, non-​linear approach, 170
Tabula Muris Atlas, 174 t-​test, 332
Index 415

Tumor mutation burden (TMB), 241 Variational Autoencoder (VAE), 168


Tumor suppressor genes, 33 VarScan2, 219
VeloCyto, 184
U Velvet, 271, 285, 347
VEP tool, 245
UCSC Genome Browser’s Variant
Viral invasion, 49
Annotation Integrator, 229
Uniform Manifold Approximation and
Projection (UMAP), 167, 170 W
UniProt, 352
Wald test, 132
Unique molecular identifiers (UMIs),
Washington University EpiGenome
153, 159, 207, 242
Browser, 331
Universal Sequencing Technology, 273
Water-​soluble substances, 7
Unix/​Linux, 109
WELLS file, 72
3’ Untranslated region (UTR), 36
Whole genome/​exome sequencing
“Unzip” process, 279
called variants, annotation of, 228–​229
Uracil-​DNA glycosylase, 240
data preprocessing, 216–​217
User-​configured computing, 107
genotyping/​variation discovery, 215
User-​friendly interface, 109
germline/​indel calling, 217–​219
U.S. NIH Roadmap Epigenomics Project,
mapping, 216–​217
322
realignment, 216–​217
recalibration, 216–​217
V
short indel calling, 217–​225
VAAST Variant Prioritizer (VVP), 247 single nucleotide variant (SNV),
VaRank, 248 217–​225
VarDict, 219 somatic mutation detection, 219–​220
VarElect, 246 structural variant, see structural
VarFish, 247 variant (SV) calling
VariantAnnotation tool, 245 variant call format (VCF), 221–​225
Variant call format (VCF), 218, 221–​225 variant calling, 221
BCF files, 223 Whole metagenome sequencing
clinical sequencing general data data normalization, 354–​355
analytic workflow, 244 experimental design/​sample
Large Scale Genomics Work Stream, preparation, 342
222 gene function annotation, 352–​353
mandatory fields, 223 gene function profiling, 353–​354
paired-​reads (PR), 225 genomic elements calling, from
read depth (RD), 225 metagenomic sequences, 351
visualization, 223 integrated metagenomics data
Variant Effect Predictor (VEP), 229 analysis, 355
Variant filtering process, 247 metabolic pathway reconstruction,
Variant Interpretation for Cancer (VIC), 353–​354
256 metagenome assembly, 347–​348
VariantMetaCaller, 224 metagenome sample collection, 343
Variant Prioritization Ordering Tool metagenome sample processing,
(VPOT), 247–​248 343–​344
Variant Ranker, 247, 248 metagenomics data repositories,
Variants, standardize classification, 248 355–​356
416 Index

microbial community analysis, Winnowmap/​Winnowmap2, 91


341–​342 Workflow Description Language (WDL),
OTUs, identify species, 355 372
sequence binning, 348–​351
sequencing approaches, 344–​345
Z
sequencing data quality control, 347
shotgun metagenome sequencing data Zebrafish Information Network (ZFIN),
analysis, 345–​346 247
taxonomic profiling, 351–​352 Zero-​inflated factor analysis (ZIFA), 167
WIG track files, 309 Zero-​inflated negative binomial (ZINB)
Wilcoxon rank sum test, 173 model, 180, 184
Window Tag Density (WTD), 307 zUMIs, 148, 154, 156

You might also like