Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Bioinformatics in MicroRNA Research 1st Edition Coll. download pdf

Download as pdf or txt
Download as pdf or txt
You are on page 1of 60

Download the full version of the ebook at

https://ebookultra.com

Bioinformatics in MicroRNA Research 1st


Edition Coll.

https://ebookultra.com/download/bioinformatics-in-
microrna-research-1st-edition-coll/

Explore and download more ebook at https://ebookultra.com


Recommended digital products (PDF, EPUB, MOBI) that
you can download immediately if you are interested.

MicroRNA in Regenerative Medicine 1st Edition Chandan K.


Sen

https://ebookultra.com/download/microrna-in-regenerative-medicine-1st-
edition-chandan-k-sen/

ebookultra.com

Essays in Bioinformatics 1st Edition S. Jelaska

https://ebookultra.com/download/essays-in-bioinformatics-1st-edition-
s-jelaska/

ebookultra.com

Advances in Bioinformatics Miguel P. Rocha

https://ebookultra.com/download/advances-in-bioinformatics-miguel-p-
rocha/

ebookultra.com

Hawaii 12th Edition Coll.

https://ebookultra.com/download/hawaii-12th-edition-coll/

ebookultra.com
Canada 13th Edition Coll.

https://ebookultra.com/download/canada-13th-edition-coll/

ebookultra.com

Japan 2017 15th Edition Coll.

https://ebookultra.com/download/japan-2017-15th-edition-coll/

ebookultra.com

Dont download 6th Edition Coll.

https://ebookultra.com/download/dont-download-6th-edition-coll/

ebookultra.com

Pattern Discovery in Bioinformatics Theory Algorithms 1st


Edition Laxmi Parida

https://ebookultra.com/download/pattern-discovery-in-bioinformatics-
theory-algorithms-1st-edition-laxmi-parida/

ebookultra.com

Integrative Cluster Analysis in Bioinformatics 1st Edition


Basel Abu-Jamous

https://ebookultra.com/download/integrative-cluster-analysis-in-
bioinformatics-1st-edition-basel-abu-jamous/

ebookultra.com
Bioinformatics in MicroRNA Research 1st Edition Coll.
Digital Instant Download
Author(s): coll.
ISBN(s): 9781493970445, 1493970445
Edition: 1st
File Details: PDF, 5.13 MB
Year: 2017
Language: english
Methods in
Molecular Biology 1617

Jingshan Huang · Glen M. Borchert


Dejing Dou · Jun (Luke) Huan
Wenjun Lan · Ming Tan · Bin Wu
Editors

Bioinformatics
in MicroRNA
Research
Methods in Molecular Biology

Series Editor:
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK

For further volumes:


http://www.springer.com/series/7651
Bioinformatics in MicroRNA
Research
Editors

Jingshan Huang
School of Computing, University of South Alabama, Mobile, AL, USA

Glen M. Borchert
Department of Pharmacology, University of South Alabama, Mobile, AL, USA;
Department of Biology, University of South Alabama, Mobile, AL, USA

Dejing Dou
Department of Computer and Information Science, University of Oregon, Eugene, OR, USA

Jun (Luke) Huan


Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA

Wenjun Lan
School of Bio-Engineering, Qilu University of Technology, Jinan, Shandong, China

Ming Tan
Mitchel Cancer Institute, University of South Alabama, Mobile, AL, USA

Bin Wu
Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
Editors
Jingshan Huang Glen M. Borchert
School of Computing Department of Pharmacology
University of South Alabama University of South Alabama
Mobile, AL, USA Mobile, AL, USA
Department of Biology
Dejing Dou
University of South Alabama
Department of Computer
Mobile, AL, USA
and Information Science
University of Oregon
Jun (Luke) Huan
Eugene, OR, USA
Department of Electrical Engineering
and Computer Science
Wenjun Lan
University of Kansas
School of Bioengineering
Lawrence, KS, USA
Qilu University of Technology
Jinan, Shandong, China
Ming Tan
Mitchel Cancer Institute
Bin Wu
University of South Alabama
Department of Endocrinology
Mobile, AL, USA
First Affiliated Hospital
Kunming Medical University
Kunming, Yunnan, China

ISSN 1064-3745     ISSN 1940-6029 (electronic)


Methods in Molecular Biology
ISBN 978-1-4939-7044-5 ISBN 978-1-4939-7046-9 (eBook)
DOI 10.1007/978-1-4939-7046-9

Library of Congress Control Number: 2017937362

© Springer Science+Business Media LLC 2017


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
express or implied, with respect to the material contained herein or for any errors or omissions that may have been made.
The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Printed on acid-free paper

This Humana Press imprint is published by Springer Nature


The registered company is Springer Science+Business Media LLC
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface

As a special class of noncoding RNAs, microRNAs (miRNAs or miRs for short) have been
reported to perform important roles in various biological and pathological processes by
regulating respective target genes. To completely understand and fully delineate miR func-
tions, besides performing biological experiments and querying PubMed and TarBase for
biologically validated miR targets, biologists can also query various miR target prediction
databases/websites for computationally predicted targets. More often than not, biologists
need to extract additional information for each and every miR target, either validated or
putative, with regard to its related information such as protein functions and affiliated sig-
naling pathways. In short, biologists are facing significant barriers in fully delineating miR
functions and the following effective bio-curation. Therefore, there is an urgent need for a
comprehensive book focusing on miR target genes, miR regulation mechanisms, miR func-
tions performed in various human diseases, and miR databases/knowledge bases.
This book is intended to give an in-depth introduction to and discussion of miRs and
their targets, miR functions, and computational techniques applied in miR research. The
primary audience includes, but is not limited to, computational biologists, computer scien-
tists, bioinformaticians, bench biologists, and clinical investigators. No prior knowledge of
computer science, databases, semantic technologies, or molecular biology is assumed. But we
do assume that readers have some biology background knowledge at the high-school level.
A brief overview of the book structure is as follows. Chapter 1 introduces the concepts of
miRs and long noncoding RNAs (lncRNAs) as well as some recent advances in miR/lncRNA
biology. Chapters 2, 3, and 4 discuss protein participants in miR regulation; viral microRNAs,
host miRs regulating viruses, and bacterial miR-like RNAs; and biomarkers, diagnostics, and
therapeutics aspects of miRs, respectively. Chapter 5 introduces basic concepts of relational
databases and biomedical big data. Chapter 6 provides an overview of semantic technologies
and bio-ontologies. Chapter 7 discusses genome-wide analysis of miR-regulated transcripts.
Chapters 8 and 9 describe in detail computational prediction of miR target genes, regulatory
interactions between miRs and their targets, as well as an introduction of various miR target
prediction databases and relevant Web resources. Chapter 10 discusses some limitations of
existing approaches that aim to improve miR target prediction accuracy. Chapters 11 and 12
introduce genomic regulation of miR expression in disease development and next generation
sequencing for miR expression profile. Chapters 13 through 16 discuss advanced topics in
computational/bioinformatics approaches in miR research, including the handling of high-
dimension data, identification and removal of noisy data, logical reasoning, and machine
learning techniques. Finally, Chapters 17–19 introduce some advances of miR research in
three human diseases: diabetes, obesity, and thyroid carcinoma.

Mobile, AL, USA Jingshan Huang


Mobile, AL, USA  Glen M. Borchert
Eugene, OR, USA  Dejing Dou
Lawrence, KS, USA  Jun (Luke) Huan
Jinan, Shandong, China  Wenjun Lan
Mobile, AL, USA  Ming Tan
Kunming, Yunnan, China  Bin Wu

v
Contents

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix

1 MicroRNAs, Long Noncoding RNAs, and Their Functions


in Human Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Min Xue, Ying Zhuo, and Bin Shan
2 MicroRNA Expression: Protein Participants in MicroRNA Regulation . . . . . . . . . . 27
Valeria M. King and Glen M. Borchert
3 Viral MicroRNAs, Host MicroRNAs Regulating Viruses,
and Bacterial MicroRNA-Like RNAs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Sara-Elizabeth Cardin and Glen M. Borchert
4 MicroRNAs: Biomarkers, Diagnostics, and Therapeutics . . . . . . . . . . . . . . . . . . . . 57
Weili Huang
5 Relational Databases and Biomedical Big Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
N.H. Nisansa D. de Silva
6 Semantic Technologies and Bio-Ontologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Fernando Gutierrez
7 Genome-Wide Analysis of MicroRNA-Regulated Transcripts . . . . . . . . . . . . . . . . . 93
David Chevalier and Glen M. Borchert
8 Computational Prediction of MicroRNA Target Genes,
Target Prediction Databases, and Web Resources . . . . . . . . . . . . . . . . . . . . . . . . . . 109
Justin T. Roberts and Glen M. Borchert
9 Exploring MicroRNA::Target Regulatory Interactions
by Computing Technologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
Yue Hu, Wenjun Lan, and Daniel Miller
10 The Limitations of Existing Approaches in Improving MicroRNA
Target Prediction Accuracy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
Rasiah Loganantharaj and Thomas A. Randall
11 Genomic Regulation of MicroRNA Expression
in Disease Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
Feng Liu
12 Next-Generation Sequencing for MicroRNA Expression Profile . . . . . . . . . . . . . . . 169
Yue Hu, Wenjun Lan, and Daniel Miller
13 Handling High-Dimension (High-Feature) MicroRNA Data . . . . . . . . . . . . . . . . . 179
Yue Hu, Wenjun Lan, and Daniel Miller
14 Effective Removal of Noisy Data Via Batch Effect Processing . . . . . . . . . . . . . . . . . 187
Ryan G. Benton
15 Logical Reasoning (Inferencing) on MicroRNA Data . . . . . . . . . . . . . . . . . . . . . . . 197
Jingsong Wang

vii
viii Contents

16 Machine Learning Techniques in Exploring MicroRNA Gene Discovery,


Targets, and Functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
Sumi Singh, Ryan G. Benton, Anurag Singh, and Anshuman Singh
17 Involvement of MicroRNAs in Diabetes and Its Complications . . . . . . . . . . . . . . . 225
Bin Wu and Daniel Miller
18 MicroRNA Regulatory Networks as Biomarkers in Obesity:
The Emerging Role . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241
Lihua Zhang, Daniel Miller, Qiuping Yang, and Bin Wu
19 Expression of MicroRNAs in Thyroid Carcinoma . . . . . . . . . . . . . . . . . . . . . . . . . . 261
Gaohong Zhu, Lijun Xie, and Daniel Miller

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281
Contributors

Ryan G. Benton • Department of Computer Science, University of South Alabama


School of Computing, Mobile, AL, USA
Glen M. Borchert • Department of Pharmacology, University of South Alabama,
Mobile, AL, USA; Department of Biology, University of South Alabama, Mobile, AL,
USA
Sara-Elizabeth Cardin • Department of Biology, University of South Alabama,
Mobile, AL, USA
David Chevalier • Department of Biology, East Georgia State College, Swainsboro,
GA, USA
N.H. Nisansa D. de Silva • Department of Computer and Information Science,
University of Oregon, Eugene, OR, USA
Fernando Gutierrez • Department of Computer and Information Science,
University of Oregon, Eugene, OR, USA
Yue Hu • College of Bioengineering, Qilu University of Technology, Jinan, Shandong,
People’s Republic of China
Weili Huang • Miracle Query, Incorporated, Eugene, OR, USA
Valeria M. King • Department of Biology, University of South Alabama, Mobile, AL, USA
Wenjun Lan • School of Bioengineering, Qilu University of Technology, Jinan,
Shandong, People’s Republic of China
Feng Liu • National Research Center for Translational Medicine (Shanghai), Rui-Jin
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
Rasiah Loganantharaj • Bioinformatics Research Lab, The Center for Advanced
Computer Studies, University of Louisiana, Lafayette, LA, USA
Daniel Miller • School of Computing, University of South Alabama, Mobile, AL, USA
Thomas A. Randall • Integrative Bioinformatics, National Institute of Environmental
Health Sciences, National Institutes of Health, Research Triangle Park, Durham,
NC, USA
Justin T. Roberts • Department of Biology, University of South Alabama,
Mobile, AL, USA
Bin Shan • Elson S. Floyd College of Medicine, Washington State University, Spokane,
WA, USA
Anshuman Singh • School of Computer Science and Mathematics, University of
Central Missouri, Warrensburg, MO, USA
Anurag Singh • Center for Advanced Computer Studies, University of Louisiana,
Lafayette, LA, USA
Sumi Singh • School of Computer Science and Mathematics, University of Central
Missouri, Warrensburg, MO, USA
Jingsong Wang • Oracle Corporation, Redwood Shores, CA, USA
Bin Wu • Department of Endocrinology, First Affiliated Hospital, Kunming Medical
University, Kunming, Yunnan, China

ix
x Contributors

Lijun Xie • Department of Nuclear Medicine, First Affiliated Hospital of Kunming


Medical University, Kunming, Yunnan, China
Min Xue • Xuzhou College of Medicine, Xuzhou, Jiangsu, China
Qiuping Yang • Department of Geriatrics, First Affiliated Hospital of Kunming
Medical University, Kunming, Yunnan, China
Lihua Zhang • Department of Geriatrics, First Affiliated Hospital of Kunming
Medical University, Kunming, Yunnan, China
Gaohong Zhu • Department of Nuclear Medicine, First Affiliated Hospital
of Kunming Medical University, Kunming, Yunnan, China
Ying Zhuo • Kadlec Regional Medical Center, Richland, WA, USA
Chapter 1

MicroRNAs, Long Noncoding RNAs, and Their Functions


in Human Disease
Min Xue, Ying Zhuo, and Bin Shan

Abstract
Majority of the human genome is transcribed into RNAs with absent or limited protein-coding potential.
microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are two major families of the non-protein-­
coding transcripts. miRNAs and lncRNAs can regulate fundamental cellular processes via diverse mecha-
nisms. The expression and function of miRNAs and lncRNAs are tightly regulated in development and
physiological homeostasis. Dysregulation of miRNAs and lncRNAs is critical to pathogenesis of human
disease. Moreover, recent evidence indicates a cross talk between miRNAs and lncRNAs. Herein we
review recent advances in the biology of miRNAs and lncRNAs with respect to the above aspects. We
focus on their roles in cancer, respiratory disease, and neurodegenerative disease. The complexity, flexibil-
ity, and versatility of the structures and functions of miRNAs and lncRNAs demand integration of experi-
mental and bioinformatics tools to acquire sufficient knowledge for applications of these noncoding
RNAs in clinical care.

Key words MicroRNA, Long noncoding RNA

1 Introduction

Majority of the human genome is transcribed although only ~2%


of the human genome encodes proteins [1]. The transcribed
RNAs with absent or limited protein-coding potential are named
noncoding RNAs and operationally divided into small RNAs and
long noncoding RNAs (lncRNA) with a boundary set at 200
nucleotides in length. The small RNA family includes microR-
NAs (miRNA), small nuclear RNAs, and piwi-interacting RNAs.
miRNAs and lncRNAs are critical regulators of development,
physiology, and disease. Herein we review recent advances in the
biology of miRNAs and lncRNAs and their functions in human
disease.

Jingshan Huang et al. (eds.), Bioinformatics in MicroRNA Research, Methods in Molecular Biology, vol. 1617,
DOI 10.1007/978-1-4939-7046-9_1, © Springer Science+Business Media LLC 2017

1
2 Min Xue et al.

2 Functions of miRNAs and Human Disease

2.1 Biogenesis miRNAs are ~22-nucleotide long single stranded RNAs that regu-
of miRNAs late gene expression via diverse mechanisms [2]. Since discovery of
the first miRNA lin-4 in Caenorhabditis elegans in 1993, 35,828
mature miRNAs have been catalogued in 223 species in the latest
release of miRBase (www.mirbase.org) [3, 4]. Biogenesis of miR-
NAs starts with transcription from a miRNA-hosting gene, which
yields a long primary transcript named primary miRNA (pri-­
miRNA) [5]. Then the pri-miRNA is cleaved by the ribonulease
III-type protein Drosha in the nucleus to produce a ~70-­nucleotide
long hairpin structure named precursor miRNA (pre-miRNA) [6].
The pre-miRNA is exported to the cytoplasm by exportin-5 and
subsequently cleaved by another ribonulease III-type protein Dicer
to generate a miRNA:miRNA* duplex of ~22 nucleodtides [7].
The miRNA:miRNA* duplex binds to an argonaute (AGO) pro-
tein to form an effector RNA-induced silencing complex (RISC)
complex. A mature miRNA is produced when miRNA* is peeled
off from the duplex. It is noteworthy that a miRNA* is not simply
a nonfunctional byproduct of miRNA biogenesis but rather a func-
tional miRNA on many occasions [8].
Besides their canonical destination in the cytoplasm miRNAs
exist and function in the nucleus and secretary microvesicles called
exosomes [9, 10]. Exosomes are small extracellular membrane ves-
icles with sizes of 30–100 nm in diameter and secreted by various
types of cells in the body [11–14]. miRNAs packaged in exosomes
can be taken up by neighboring cells or distant recipient cells via
transportation in body fluids and function in their recipient cells,
which serve as an important tool for proximal and distant intercel-
lular communications [15–18].
Biogenesis of miRNAs can be regulated at every step of their
production by physiological and pathological signals. For instance
the miRNA-200 family is transcriptionally suppressed by ZEB1
during epithelial–mesenchymal transition (EMT) [19]. In another
example type I collagen posttranscriptionally upregulates the
expression of miR-21 by promoting maturation of pre-miR-21 to
miR-21 without alteration in the amount of pri-miRNA-21 and
pre-miR-21 [20].

2.2 Functions The classic mode of a miRNA’s action is to inhibit gene expression
of miRNA via binding to its complementary sequences (6–8 nucleotides)
within the 3′ untranslated region (3′ UTR) of its target mRNAs.
This partial complementarity causes inhibition of expression of a
miRNA’s target via degradation or repression of translation of the
bound mRNAs [21]. Because of the need of only a 6–8 nucleotide
complementarity a miRNA can potentially targets hundreds of
mRNAs and most mammalian mRNAs are conserved targets of
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 3

miRNAs [22]. Bioinformatic tools such as TargetScan have been


widely used to guide prediction and validation of a miRNA’s target
mRNAs [23].
The cytoplasmic miRNAs inhibit their target genes expression
via degradation of mRNAs or inhibition of translation at initiation
and post-initiation steps [21, 24–31]. miRNA-mediated decrease
of their target mRNA levels is proposed as a major mechanism of
miRNA-mediated repression of a target gene expression [32]. This
action can be achieved through miRNA-induced rapid deadenyl-
ation of target mRNA as exemplified by the actions of miR-125b,
a miRNA that is linked to chemoresistance in breast cancer [31,
33, 34]. miRNA-mediated suppression of translation initiation is
exemplified in let-7-mediated repression of its target mRNAs as
let-7-bound AGO2 represses the translation initiation by binding
to the m7G cap of the mRNA targets and thereby prevents the
recruitment of eIF4E, an essential translation initiation factor [35].
The post-initiation inhibition of translation by miRNAs is accom-
plished through rapid degradation of the peptide product encoded
by the targeted mRNA, which is mediated by high rate of ribo-
some drop-off during translation elongation [36].
In addition to their canonical actions in the cytoplasm miR-
NAs are expressed in abundance in the nucleus and regulate vari-
ous nuclear events such as transcription and RNA splicing via
diverse mechanisms. For instance miR-320 recruits AGO1 and
EZH2 to the POLR3D locus through complete complementary
binding, which results in heterochromatinization and silencing of
the POLR3D promoter [37]. miRNA-mediated silencing of the
gene promoters harboring their target sites controls a variety of
fundamental cellular processes, such as cellular senescence and
neuroregeneration [38–40]. On the other hand, a few nuclear
miRNAs have been reported to activate gene expression via epi-
genetic mechanisms. For instance miR-373 activates the expres-
sion of CDH1 and CSDC2 via AGO-miRNA complexes-mediated
recruitment of positive epigenetic regulators to the target promot-
ers [41]. Lastly nuclear miRNAs can regulate splicing of the com-
plement pre-mRNA. In miR-122-mediated repression of splicing
of the hepatitis C viral RNA a ternary complex formed between the
target transcript, miRNA, and RISC masks splicing recognition
motifs and thereby prevents binding of the splicing factors [42].

2.3 miRNAs miRNAs govern fundamental biological processes, such as cell pro-
and Human Disease liferation, death, differentiation, and development [43]. As a
­feedback tool with profound effects on gene expression miRNAs
are the main tool to fine-tune gene expression and biological
homeostasis. Dysregulation of miRNAs contributes to pathogen-
esis of a wide variety of human disease. In this section we review
actions of miRNAs in cancer, respiratory disease, and neurodegen-
erative disease.
4 Min Xue et al.

2.3.1 miRNAs in Cancer The first documented association between miRNAs and cancer is
frequent deletion and downregulation of miR-15 and miR-16 at
13q14 in chronic lymphocytic leukemia [44]. Since then, thou-
sands of miRNAs have been reported to act as either oncogenes or
tumor suppressors depending on a miRNA’s targets in a particular
biological context. miRNAs have been linked to each hallmark of
cancer that is established by Hanahan and Weinberg [45].
Representative miRNAs associated with each hallmark of cancer
are listed in Table 1 [46–62].
Genetic alterations are a common cause of dysregulation of
miRNAs in cancer. More than 50% of miRNA genes are located in
cancer associated genomic regions or in fragile sites [63]. One
prime example is amplification of the oncogenic miR-17~92 clus-
ter and its consequent overexpression in small cell lung cancer
[47]. Deletion and loss of expression of miRNAs in cancer are
exemplified in frequent deletion of the miR-15a and miR-16a

Table 1
Association between miRNA and hallmarks of cancer

Hallmarks of cancer Representative miRNAs


Sustaining proliferative signaling miR-17~92-mediated suppression of PTEN in lung cancer and
B-cell lymphoma [46, 47]; Loss of let-7-­mediated suppression of
Ras by in lung cancer [48, 49]
Evading growth suppressors Interference of cell cycle arrest by miR-675-mediated suppression
of pRB in colorectal cancer [50]
Avoiding immune destruction Enhancement of resistance to cytotoxic T-lymphocytes by
miR-222 mediated suppression of ICAM-1 [51]
Enabling replicative immortality Loss of miR-34a-mediated senescence in colon cancer [52]
Tumor promoting inflammation miR-155-mediated inflammation in the tumor microenvironment
[53, 54]
Activating invasion and metastasis miR-10b-mediated migration, invasion, and metastasis in breast
cancer [62]; Loss of miR-200-mediated suppression of EMT
[55, 56]
Inducing angiogenesis Enhanced angiogenesis by miR-296-mediated suppression of
HGS in tumor associated endothelial cells in gliomas [57]
Genome instability and mutation Impairment of DNA repair by miR-21-mediated suppression of
H2AX, a histone variant essential to repair [58, 59]
Resisting cell death Inhibition of caspase activation by miR-21-­mediated suppression
of PDCD4 in glioblastoma [60]
Deregulating cellular energetics Loss of miR-99a/100-­mediated suppression of mTOR in
childhood adrenocortical tumors [61]
A summary of representative miRNAs associated with the hallmarks of cancer. PTEN phosphatase and tensin homolog,
pRB retinoblastoma protein, EMT epithelial–mesenchymal transition, HGS hepatocyte growth factor-regulated tyrosine
kinase substrate, PDCD4 programmed cell death protein 4, mTOR mechanistic target of rapamycin
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 5

hosting locus in chronic lymphocytic leukemia [44]. Single nucle-


otide polymorphism (SNP) is another common cause of dysregula-
tion of miRNAs in cancer. In a common G/C polymorphism
(rs2910164) within the pre-miR-146a coding region the C allele
results in a decrease of mature miR-146a and less efficient inhibi-
tion of the miR-146a targets, which increases risk of papillary thy-
roid carcinoma [64]. Variation within the miRNA target site of a
miRNA-targeted 3′UTR is another important source of genetic
predisposition in cancer risk. In the oncogenic HMGA2 locus the
open reading frame and the 3′UTR harboring the let-7 target sites
are separated by chromosomal rearrangements in cancer, which
leads to escape of HMGA2 from let-7-mediated repression [65, 66].
SNP in a miRNA target site can result in loss of miRNA-­mediated
repression in cancer. In the let-7 target site within the 3′UTR of
the KRAS oncogene SNP causes elevated KRAS expression and
increased risk of non-small cell lung cancer [67].
Transcriptional dysregulation of miRNA expression is another
critical mechanism in tumorigenesis. The oncogenic miRNAs are
often transcriptionally activated in cancer [68]. For instance the
oncogenic miR-17~92 cluster is transcriptionally activated by the
MYC oncogene via a MYC binding site in the promoter of the
miR-17~92 cluster [69]. In contrast the tumor suppressive miR-
NAs are often transcriptionally repressed in cancer [70]. The pro-
moter of the miR-200 cluster that encodes miR-200a, miR-200b,
and miR-429 is transcriptionally repressed by ZEB1 and SIP1 dur-
ing EMT, a process through which cancer cells acquire invasive
and metastatic competency [19, 55]. More importantly the miR-­
200 cluster members repress the expression of ZEB1 and SIP1 via
the miR-200 target sites in their 3′ UTR and this reciprocal repres-
sion between the miR-200 cluster and ZEB1/SIP1 establishes a
double-negative feedback loop in regulation of EMT [19, 55].
miRNA expression can also be regulated by the signals from the
tumor microenvironment, such as extracellular matrix, and in turn
mediates cancer cell’s responses to the tumor microenvironment
[20, 56, 71, 72].
Because of the critical roles of miRNAs in cancer biology miR-
NAs have emerged as a family of promising targets in diagnosis and
treatment of cancer. Because miRNAs are more stable than mRNAs
and released by a solid tumor into the body fluids via exosomes
miRNAs have emerged as promising biomarkers in tissue biopsies,
blood, urine, etc. [73–75]. For instance a host of circulating miR-
NAs including miR-141, miR-21, and miR-92a have been tested
as diagnostic biomarkers of colorectal cancer in whole plasma or
serum [76–81]. miRNAs have also been developed as molecular
signatures of subtypes of breast cancer and thus guide the treat-
ment that is tailored for each molecular subtype. The miRNA sig-
natures of ER+ and HER+ can guide anti-ER and anti-HER2
therapies, respectively [82–84]. miRNAs can potentially predict
6 Min Xue et al.

responses to chemotherapy and thus guide the choice of treat-


ments as illustrated in the miRNA signatures that can predict
response to tamoxifen and anti-HER2 monoclonal antibody
Herceptin in breast cancer [85–87].
Current development of miRNA-based therapies mainly
employs antagonist and oligonucleotide mimics of a miRNA of
interest. miRNA mimics are used to restore tumor suppressive
miRNAs that are deficient in cancer. On the contrary antagomiRs
are single-stranded oligonucleotides that complement and inhibit
the oncogenic miRNAs in cancer. To increase efficiency of a
miRNA antagonist, miRNA sponge technology has been ­developed
to synthesize a single stranded RNA containing multiple binding
sites of a targeted miRNA to efficiently neutralize a miRNA [88].
miRNA sponges have been validated in xenograft mouse model of
human breast cancer cell lines in that inhibition of miRNA-9 and
miR-150 using a synthetic RNA containing several miR-9 or miR-
150 binding sites reduced lung metastases [89, 90]. The miRNA
targeting therapies have entered clinical trials as exemplified by a
miR-34a mimics in phase I study (http://clinicaltrials.gov/ct2/
show/NCT01829971). Preliminary results from the translational
studies of miRNA-based therapies against cancer suggest that
miRNA mimics or antagomiRs can be easily administered through
local or parenteral injection routes with sufficient uptake of the
agents to achieve sustained and desired effects in the targeted tis-
sues and organs.

2.3.2 miRNAs miRNAs have emerged as critical regulators in the control of nervous
in Neurodegenerative system-specific gene expression during development, aging, and dis-
Disease ease. We review the role of miRNAs in two devastating neurodegen-
erative diseases, Parkinson’s disease and Alzheimer’s disease.
Parkinson’s disease is a chronic and progressive movement dis-
order that is caused by a gradual loss of midbrain dopaminergic
neurons [91]. Investigation of miRNAs has shed light on patho-
genesis of Parkinson’s disease. miR-133b is specifically expressed in
the midbrain dopaminergic neurons and regulates maturation and
function of the midbrain dopaminergic neurons as a node of a neg-
ative feedback circuit by targeting the paired-like homeodomain
transcription factor Pitx3 [92]. Importantly, miR-133b is deficient
in the midbrain tissues from patients with Parkinson’s disease [92].
Gain-of-function mutations in leucine-rich repeat kinase-2
(LRRK2) cause familial and sporadic Parkinson’s disease. The
pathogenic LRRK2 associate with RISC to interfere the miRNA
pathway and such interference leads to overproduction of E2F1/DP,
a target of let-7 and miR-184* [93]. Moreover, antagomiR-­
mediated blockage of let-7 or miR-184* can recapitulate the toxic
effects of the pathogenic LRRK2 and conversely forced expression
of let-7 or miR-184* can attenuate the toxic effects of the patho-
genic LRRK2 [93].
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 7

Alzheimer’s disease is the most common cause of dementia in


the aging population. The pathologic hallmarks of the disease are
plaques composed of amyloid β and tangles composed of hyper-
phosphorylated tau [94]. Several miRNA profiling studies have
identified dozens of miRNAs that are significantly differentially
expressed between cortex from patients with Alzheimer’s disease
and the matching controls [95–97]. The differentially expressed
miRNAs appear to be more robust and reproducible than the dif-
ferentially expressed mRNAs [98]. Moreover, a joint profiling of
miRNA and mRNA expression in brain cortex from Alzheimer’s
disease and age-matched control subjects reveals strong inverse
correlations between the amount of miRNAs and their corre-
sponding predicted mRNA targets, which suggests active
microRNA functions in Alzheimer’s disease [99]. Indeed the
expression of miR-29a, miR-29b-1, and miR-9 is significantly
decreased in Alzheimer’s disease and their decrease causes aberrant
increase of their target β-secretase-1 (BACE1), a protein account-
able for accumulation of Aβ in Alzheimer’s disease [100]. Besides
BACE1, the expression of amyloid precursor protein is repressed
by the members of the miR-20a family (miR-20a, miR-17-5p, and
miR-106b) and this miRNA pathway appears to be compromised
in Alzheimer’s disease because miR-106b is substantially reduced
in sporadic Alzheimer’s disease [101].

2.3.3 miRNAs As in many other tissues miRNAs mediate cell differentiation and
in Respiratory Disease maintain homeostasis in differentiated cells in the respiratory sys-
tem [102]. miRNA expression undergoes profound changes dur-
ing lung development and the Dicer null mice are not viable due
to impaired lung growth that is caused by deficiency in production
of mature miRNAs globally due to deletion of Dicer [103, 104].
Moreover, expression of the miR-17~92 cluster progressively
declines during lung development and forced expression of the
cluster results in abnormal lung development that was character-
ized by continued proliferation and impaired differentiation of epi-
thelial cells [105].
Profound alteration in miRNA expression profile has been
observed in asthma and asthma undergoing steroid therapy [106,
107]. More importantly the altered miRNA expression profile
observed in asthma appears to be largely driven by IL-13, a key
pathogenic cytokine in asthma because the miRNA profile in
asthma can be recapitulated by exposing lung epithelial cells to
IL-13 [107]. T cells are important orchestrators of the chronic
inflammatory response in asthma. In the circulating CD4+ and
CD8+ T cells collected from the patients with severe asthma the
expression of miR-146a and miR-146b are substantially downreg-
ulated and such downregulation potentially contributes to greater
T-cell activation in severe asthma because these two miRNAs
inhibit the immune response [108, 109].
8 Min Xue et al.

Idiopathic pulmonary fibrosis is a deadly lung disease featuring


excessive production and deposition of extracellular matrix com-
ponents by fibroblasts and myofibroblasts in the lung interstitium.
miRNA expression profiling of human lung samples, isolated lung
cells, and mouse models of idiopathic pulmonary fibrosis have
revealed profound dysregulation of miRNA that includes let-7d,
miR-21, and miR-29 [110–114]. More importantly reversing the
expression pattern of let-7d and miR-21 observed in pulmonary
fibrosis can attenuate pulmonary fibrosis in mice, suggesting that
these miRNAs mediate fibrosis in the lung [110–114]. It is note-
worthy that miR-29 represses expression of a host of extracellular
matrix proteins and has been implicated as a key regulator in
fibrotic diseases in other organs as well [115].
Chronic obstructive pulmonary disease (COPD) is caused pre-
dominantly by long-term exposure to cigarette smoke and charac-
teristic of destruction of the lung parenchyma (emphysema) and
reduced lung function. A miRNA expression profiling of lung tis-
sues collected from smokers with and without COPD has revealed
a panel of 70 miRNAs that are differentially expressed between
smokers with and without COPD [116]. This panel is enriched
with the miRNAs linked to the pathways that underlie the patho-
genesis of COPD, such as the TGF-β, Wnt, and focal adhesion
pathways [116]. For instance upregulated expression of miR-15b
is observed only in the smoker with COPD and correlated with a
decrease of its validated target SMAD7, a well-established inhibitor
of the TGF-β pathway [116]. In two other in-depth studies
reduced miR-146a expression is linked to increased expression of
PGE2 due to loss of miR-146a-mediated repression of COX-2 and
reduced expression of miR-1 is linked to the muscle weakness
observed in COPD [117, 118].

3 Functions of lncRNAs and Human Disease

lncRNA is a heterogenous family that is represented by long inter-


genic RNAs (defined by position), circular RNAs (circRNA,
defined by structure), competing endogenous RNA (ceRNA,
defined as functions), antisense RNAs (defined by orientation of
transcription), etc. According to the Ensembl human genome
annotation (GRch38, version 23) 27,817 lncRNAs are transcribed
from 15,931 gene loci [119]. lncRNAs are of absent or limited
protein coding potential. However, increasing number of genes
can dually produce peptides/proteins and lncRNAs. For example,
the RNA and proteins products of the steroid receptor RNA acti-
vator gene are simultaneously produced and the RNA product
function as a scaffold for formation of several ribonucleoprotein
complexes [120]. Similar to mRNA, most lncRNAs are transcribed
by RNA polymerase II, 5′-capped, 3′-polyadenylated, and spliced
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 9

into various isoforms although they tend to have fewer exons than
mRNAs [1].
The importance of lncRNA genes are revealed by their prox-
imity to developmental regulators in the genome, enrichment of
tissue-specific and developmental stage-specific expression pat-
terns, and frequent association with genetic traits [121]. lncRNAs
regulate a myriad of molecular and cellular processes, such as
­chromatin remodeling and RNA splicing [122–126]. In the fol-
lowing sections we discuss the functions of lncRNAs and their role
in human disease.

3.1 Functions of A large number of lncRNAs can recruit chromatin remodeling


lncRNAs complexes to a specific set of genes to activate or repress gene
expression [127–133]. As many as 20% of lncRNAs expressed in a
3.1.1 Regulation of
given cell associate with chromatin remodeling complexes and
Epigenetic Modifications
lncRNAs are commonly bound by multiple chromatin remodeling
proteins [134]. One of the best-characterized interactions between
lncRNAs and chromatin remodeling complexes is lncRNA-­
mediated recruitment of polycomb repressive complex 2 (PRC2)
that catalyzes trimethylation of histone H3 lysine 27 (H3K27me3),
a histone code for transcriptional repression [135].
lncRNAs can act in cis to recruit chromatin remodeling com-
plexes to regulate gene expression, which is well-characterized in
X-chromosome inactivation by the lncRNA X-inactive-specific
transcript (XIST) [136, 137]. In mammalian females, the majority
of genes on one of the two X-chromosomes in each cell are silenced.
During female development, XIST is transcribed from the
X-chromosome that is destined to become inactivated in each cell.
XIST then coats the regions of the chromosome that is to be
silenced, which results in formation of “XIST clouds” and recruit-
ment of PRC2 to the XIST-coated region for gene silencing [138].
On the other hand, lncRNAs can act in trans to recruit PRC2
to repress gene expression. As a paradigm of such mechanism, the
lncRNA HOX antisense intergenic RNA (HOTAIR) is transcribed
from the locus between HOXC11 and HOXC12 on chromosome
12 and repress the HOXD gene cluster on chromosome 2 via
recruitment of PRC2 [129]. As demonstrated by chromatin isola-
tion by RNA purification HOTAIR preferentially occupies a
GA-rich DNA motif to recruit PRC2 and nucleate broad domains
of H3K27me3 [139].
lncRNAs can regulate another important epigenetic code,
DNA methylation, a dynamic and reversible process that governs
gene expression during development and disease. In general cyto-
sine methylation in a CpG island located in a gene promoter marks
a gene for repression. An antisense lncRNA named TCF21 anti-
sense RNA inducing demethylation (TARID) activates TCF21
expression by inducing promoter demethylation [140]. TARID
recruits growth arrest and DNA-damage-inducible-α (GADD45A)
10 Min Xue et al.

to the TCF21 promoter. GADD45A, a regulator of DNA demeth-


ylation, in turn recruits thymine-DNA glycosylase for base excision
repair-mediated demethylation in the TCF21 promoter [140]. In
summary lncRNA can regulate the epigenetic codes through phys-
ical interaction with chromatin or nucleotide modifiers.

3.1.2 Regulation of RNA Besides regulation of epigenetic codes a large number of lncRNAs
Splicing regulate RNA splicing. For instance the lncRNA metastasis-­
associated lung adenocarcinoma transcript 1 (MALAT1) regulates
alternative splicing through its interaction with the serine/arginine-­
rich (SR) family of nuclear phosphoproteins, a key component of
the splicing machinery [141, 142]. MALAT1 is required for appro-
priate splicing because depletion of MALAT1 results in increase of
mislocalized and unphosphorylated SR proteins as well as increase
of exon inclusion events [142]. It is proposed that MALAT1 serves
as a structural docking site for accumulation and assembly of spe-
cific splicing factors, such as phosphorylated SR proteins and this
process is essential for efficient splicing [141].

3.1.3 Regulation Another step of a RNA life cycle regulated by lncRNAs is mRNA
of mRNA Decay decay as illustrated in staufen-1-mediated mRNA degradation
[143]. Staufen-1 binds to translationally active mRNAs via imper-
fect base-pairing between one Alu element in the 3′ UTR of a
staufen-1 target and another Alu element in a cytoplasmic, polyad-
enylated long noncoding RNA (lncRNA) [143]. Formation of the
mRNA-lncRNA-staufen-1 hetero triplex leads to degradation of
the staufen-1-targeted mRNAs [143]. It is noteworthy that this
process assigns a novel function to Alu, an ancient DNA repetitive
element, which is echoed in a novel function of another repetitive
element SINEB2 as discussed below.

3.1.4 Regulation lncRNAs can regulate the final step of an mRNA life cycle, transla-
of mRNA Translation tion, through base-pairing with the targeted mRNAs and forma-
tion of this RNA-RNA duplex that in turn modulates the interaction
between the translated mRNAs and ribosomes [144, 145]. The
antisense lncRNAs that complement the targeted mRNAs at the 5′
end promote association of polysomes with the targeted mRNAs.
A nuclear-enriched lncRNA antisense to mouse ubiquitin carboxy-­
terminal hydrolase L1 (Uchl1) can increase UCHL1 mRNA trans-
lation, which requires the presence of a 5′ overlapping sequence
and an embedded inverted SINEB2 element [145]. On the con-
trary, lincRNA-p21 can associate with JUNB mRNA and selec-
tively reduce its translation when lincRNA-p21 is released from
HuR, a RNA-binding protein that binds to and limits the avail-
ability of lincRNA-p21 for targeting JUNB [144].

3.1.5 Molecular Scaffold lncRNAs can serve as molecular scaffolds for assembly and posi-
for Structural/Functional tioning of structural or functional complexes so that the lncRNA
Complexes containing complexes can function in an appropriate spatial and
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 11

temporal manner [134, 146, 147]. HOTAIR provides a paradigm


of how lncRNAs act as a modular scaffold. HOTAIR interacts with
PRC2 and lysine-specific demethylase 1 (LSD1) corepressor com-
plex to silence the HOXD locus in trans [148]. Assembly and posi-
tioning of a transcriptional corepressor complex containing PRC2
and LSD1 on a HOTAIR bound gene promoter can efficiently
coordinate histone codes for gene silencing via PCR2-mediated
addition of repression code H3K27me3 and LSD1-mediated
removal of activation code H3K4me3. Such coordination is
achieved through scaffolding by HOTAIR in that HOTAIR binds
to PRC2 via its 5′ terminus and to LSD1 via its 3′ terminus [148].

3.2 lncRNAs lncRNAs dictates fundamental cellular and developmental pro-


and Human Disease cesses and thereby underlie pathogenesis of a broad range of
human diseases. Aside from their well-established roles in cancer,
lncRNAs are central to fragile X syndrome, neurodegenerative dis-
ease, respiratory disease, etc. [149–154]. In this section we review
recent advances on the role of lncRNAs in cancer, neurodegenera-
tive disease, and respiratory disease.

3.2.1 lncRNAs in Cancer lncRNAs have emerged as novel master regulators of initiation,
progression, and response to therapy in a wide variety of solid
tumors and hematological malignancies [155]. Hundreds of
IncRNAs are differentially expressed between tumor tissues and
paired adjacent nontumor tissues in various types of cancer [156–
164]. lncRNAs can act as oncogenes or tumor suppressors to regu-
late cancer biology via diverse molecular mechanisms [141, 158,
159, 165–167].
One of the classical and versatile cancer-associated lncRNAs is
MALAT1. Elevated expression of MALAT1 has been reported in a
broad range of cancers, including lung cancer and breast cancer
[168–184]. Moreover, genetic alterations in MALAT1, such as
multiple mutations and deletions within the SRSF1-binding sites
are associated with poor patient outcome in breast cancer [185].
In colorectal cancer and osteosarcoma, MALAT1 promotes tumor
growth and metastasis by binding to a multifunctional RNA-­
binding protein, PSF via a motif in its 3′region [186, 187].
Depletion of MALAT1 results in defective alternative splicing of a
subset of transcripts that are involved in cancer such as tissue factor
and endoglin [188]. Besides regulation of splicing of the cancer-­
associated genes MALAT1 regulates expression of the cancer asso-
ciated genes via epigenetic mechanisms. For example, MALAT1
binds to polycomb group proteins to facilitate assembly of multiple
corepressors/coactivators and thereby mediates activation of the
growth-control gene program for proliferation [189]. MALAT1
also associates with PRC2 and alters the expression of N-cadherin
and E-cadherin to promote EMT in bladder cancer cells [175].
Besides its potential as a biomarker, MALAT1 is an appealing
12 Min Xue et al.

therapeutic target for cancer metastasis because deletion or inhibi-


tion of MALAT1 reduces tumor growth and metastasis of human
lung cancer cells in xenografted mice [187].
Another prime example of oncogenic lncRNAs is HOTAIR
[190, 191]. HOTAIR is elevated in various cancers including
breast cancer, lung cancer, colorectal cancer, and prostate cancer
[159, 192–194]. Elevated HOTAIR expression is a powerful pre-
dictor of metastasis and poor survival [159]. The paradigm of
HOTAIR’s functions in cancer is that increased amount of
HOTAIR reprograms global gene occupancy of PRC2, which
results in de novo repression of hundreds of new target genes to
promote invasion and metastasis [159]. HOTAIR mediates cancer
cells’ resistance to chemotherapy because elevated expression of
HOTAIR is correlated with tamoxifen resistance in breast cancer
and its overexpression promotes ligand-independent proliferative
activities of estrogen receptor [195].
Similar to miRNAs lncRNAs can regulate the hallmarks of can-
cer via diverse mechanisms as illustrated in regulation of apoptosis
and proliferation by the lncRNA growth arrest-specific transcript 5
(GAS5). GAS5 is induced upon growth arrest due to lack of nutri-
ents or growth factors and overexpression of GAS5 induces growth
arrest and apoptosis in cancer cells [196]. Acting as molecular
decoy through its binding to the DNA-binding domain of gluco-
corticoid receptor GAS5 sequesters glucocorticoid receptor and
thereby suppresses glucocorticoid-activated genes, particularly the
inhibitors of apoptosis [196]. Another example of lncRNA-­
mediated control of cell cycle control is lincRNA-p21 whose
expression is induced by p53 upon DNA damage [166]. Upon
induction by p53 lincRNA-p21 interacts with ribonucleoprotein K
and act in trans to recruit ribonucleoprotein K to repress a host of
p53 targeted genes, particularly the genes that interfere with the
apoptotic response to DNA damage. On the other hand, lincRNA-
­p21 can function in cis to activate expression of its neighboring
gene, particularly p21, an essential mediator of p53-dependent
growth arrest response to DNA damage [197].
Given their pivotal role in cancer biology lncRNAs have
emerged as promising targets for diagnosis and therapy of cancer.
Some lncRNAs are potential biomarkers of a broad range of can-
cer, such as HOTAIR that is upregulated in the majority of can-
cers investigated so far [191]. More importantly elevated
expression of HOTAIR and MALAT1 is valuable in prognosis
because their aberrant expression correlates significantly with
metastasis and poor overall survival in breast, lung, and colorectal
cancers [160, 191, 198, 199]. On the other hand, some lncRNAs
are dysregulated in cancer in a tissue type-specific manner. For
instance the lncRNA prostate cancer antigen 3 (PCA3) is upregu-
lated ­specifically in prostate tumor tissue over normal/nonmalig-
nant tissue [200, 201].
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 13

Similar to miRNAs altered expression of lncRNAs in various


body fluids is often congruent to their dysregulated expression in
the tumor tissues. Thus acquisition of lncRNA based biomarkers in
body fluids is convenient, minimally invasive, and cost-effective
relative to conventional tissue biopsies. Particularly the circulating
lncRNAs exist in exosomes released by cancer cells because the
lncRNAs packaged into exosomes appear to be tightly controlled
and thus provide pivotal information of their parental cancer cells
that otherwise requires conventional biopsies to obtain [202, 203].
One of the most explored methods to inhibit upregulated
oncogenic lncRNAs is RNA interference. siRNAs targeting
lncRNAs via base-pairing have yielded promising efficiency in
reducing lncRNA expression and cancer growth/metastasis as
observed in targeting HOTAIR in breast cancer cells [159, 204].
Another base-pairing approach is using longer antisense oligonucle-
otides to promote degradation of the targeted lncRNA by RNase H
that was successfully applied to target MALAT1 in lung cancer cells
[168]. On the other hand, the expression of the tumor suppressive
lncRNAs may be enforced/introduced by delivery of an lncRNA
transgene. Because lncRNAs function through physical interactions
with protein partners in normal and cancer contexts one ideal
approach to specifically interfere the lncRNAs’ functions in cancer
is to disrupt the cancer-specific interaction between lncRNAs and
their protein partners. This approach has been attempted in a feasi-
bility test that successfully identified the compounds that can dis-
rupt the interaction between HOTAIR and PRC2 [205].

3.2.2 lncRNAs lncRNAs have emerged as orchestrators of gene regulatory net-


in Neurodegenerative works in the nervous system. Hundreds of lncRNAs exhibit tempo-
Disease ral and spatial patterns of expression in the nervous system, which
suggest that they play key roles in development and normal func-
tions of the nervous system [206]. As validated by a profiling and
functional analysis of lncRNAs in human embryonic stem cells effi-
cient neuronal differentiation of embryonic stem cells requires sev-
eral lncRNAs, such as lncRNA_N1 (AK124684) and lncRNA_N2
(AK091713) [207]. We review the role of lncRNAs in two neuro-
degenerative diseases, Alzheimer’s disease and Parkinson’s disease.
One salient example of lncRNAs linked to pathogenesis of
Alzheimer’s disease is the lncRNA named antisense transcript of
β-secretase-1 (BACE1-AS). BACE1 is believed to be the culprit of
accumulation of β-amyloid and the consequent formation of amy-
loid plaques that are pathological hallmarks of Alzheimer’s dis-
ease. BACE1-AS can increase the mRNA levels of BACE1 by
duplexing with and stabilizing the BACE1 mRNA [149]. More
importantly BACE1-AS is elevated in the brains of patients with
Alzheimer’s disease, suggesting that BACE1-AS mediates upregu-
lated expression of BACE1 in Alzheimer’s disease [208]. The
amount of BACE-1AS in the brains of patients with Alzheimer’s
14 Min Xue et al.

is proportional to the severity of dementia [149]. Another lncRNA


linked to Alzheimer’s disease belongs to a family of the brain cyto-
plasmic (BC) lncRNAs. The lncRNA BC200 is transported to
dendritic processes via ribonucleoprotein particles and regulate
gene expression at the translational level [209]. BC200 declines in
the frontal cortex of normal aging brain, but increases in
Alzheimer’s disease, and its increase is congruent to the severity of
dementia [210].
In Parkinson’s disease, aberrant expression of PTEN induced
kinase 1 (PINK1) leads to abnormal mitochondrial morphology,
impaired dopamine release, and motor deficits [211]. The lncRNA
natural antisense RNA of PINK1 (naPINK1) is transcribed as an
antisense transcript from the PINK1 locus and stabilizes the expres-
sion of a PINK1 splice variant (svPINK1) containing a domain
homologous to the C-terminus regulatory domain of PINK1
[212]. Silencing of naPINK1 results in decrease of svPINK1 in
neurons, which suggests that naPINK1 and svPINK1 are concor-
dantly regulated during impairment of mitochondrial biogenesis
related to Parkinson’s disease [212].

3.2.3 lncRNAs Hundreds of lncRNAs are expressed in a developmental stage-­


in Respiratory Disease specific and cell type-specific manner in the mouse lung [213].
Moreover, the lncRNA expressing genomic loci are enriched with
the binding sites for the established master transcriptional regula-
tors of lung development, such as serum response factor, forkhead
box, and SP1. In particular, two lncRNAs, NANCI and LL34,
regulate expression of the genes that govern the key steps in dif-
ferentiation and development of airway epithelial cells [213].
These findings implicate a critical role of lncRNAs in lung develop-
ment, function, and disease.
The importance of lncRNAs in respiratory disease is high-
lighted in characterization of the deletion of the locus harboring
several lncRNAs in a rare lethal neonatal lung disorder alveolar
capillary dysplasia with misalignment of pulmonary veins (ACD/
MPV) [214]. The lung-abundant 16q24.1 lncRNAs transcribed
from the deleted locus of ACD/MPV may contribute to long-­
range regulation of FOXF1 by GLI2 and other transcription fac-
tors via chromatin looping [214]. Loss of those lncRNAs due to
deletion results in loss of FOXF1 expression as well as consequent
vascular pathology observed in ACD/MPV. It is conceivable that
similarly dysregulated lncRNA expression and function occur in
pulmonary vascular remodeling in pulmonary arterial hyperten-
sion and asthma. Indeed in asthma and COPD inhibition of
MALAT1 appears to be a promising approach to attenuate occlu-
sive lesions in pulmonary arterial hypertension, inhibit airway epi-
thelial cell proliferation, and reduce obstructive remodeling of the
airways [215].
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 15

4 Regulation of miRNAs by lncRNAs

A new frontier in the noncoding RNA world is the cross talk


between miRNAs and lncRNAs [216]. Recent studies have dis-
covered and characterized naturally occurring microRNA sponges,
termed competing endogenous RNAs (ceRNAs) [217]. These
ceRNAs consist of a variety of RNA species that include protein-­
coding mRNAs, pseudogenes, and lncRNAs (including cir-
cRNAs). We focus on regulation of miRNAs by lncRNAs.
One of the well-charaterized ceRNAs is linc-RoR that is abun-
dantly expressed in human embryonic stem cells [218]. linc-RoR
sequesters miR-145 and thus protects OCT4, SOX2, and NANOG
from miR-145-mediated repression [218]. The linc-ROR-­
mediated interference of miR-145 is essential to renewal of embry-
onic stem cells [218]. In a similar fashion HOTAIR antagonizes
several tumor suppressive miRNAs. In gastric cancer cells,
HOTAIR acts as a ceRNA to trap miR-331-3p through a comple-
mentary target site in its sixth exon and consequently increases the
expression of the miR-331-3p-targeted oncogene HER2 [219].
In gall bladder cancer, HOTAIR’s oncogenic activity requires its
binding to and titration of miR-130a through a target site in its
sixth exon [220]. Interestingly the interaction between HOTAIR
and miR-130a is reciprocal because miR-130a represses the
expression of HOTAIR in a target site-dependent manner [220].
The oncogenic lncRNA H19 acts as a ceRNA of two tumor sup-
pressive miRNAs, miR-141 and let-7 and thus promotes the pro-
liferative and invasive phenotypes of cancer cells [221, 222].
Besides cancer ceRNAs play a critical role in normal physiology.
For instance linc-MD1 is a ceRNA that mediates skeletal muscle
differentiation by titrating away miR-133 and miR-135 from their
targets MAML1 and MEF2C mRNAs [223]. Decreased expres-
sion of linc-MD1 is believed to mediate pathogenesis of Duchenne
muscular dystrophy, a devastating muscle degenerative disease due
to uncontrolled repression of MAML1 and MEF2C by miR-133
and miR-135 [223].
circRNAs are structurally distinct in that they form a cova-
lently closed continuous loop in which the 3′ and 5′ ends that are
normally exposed in an linear RNA molecule are joined together
in circRNAs. Several circRNAs containing multiple target sites of
an individual microRNA have been characterized, which indicate
that circRNAs can efficiently sequester microRNAs [224]. For
instance, the circRNA sponge for miR-7, ciRS-7, contains dozens
of miR-7 target sites and is enriched in the human and mouse
brain [225]. More importantly, ciRS-7 regulates brain develop-
ment by titrating miR-7 [225].
16 Min Xue et al.

5 Conclusions

miRNAs and lncRNAs have emerged as central players in funda-


mental cellular processes, development, and pathogenesis of
human disease. The complexity, flexibility, and versatility of the
structures and functions of miRNAs and lncRNAs demand inte-
gration of experimental and bioinformatic tools to acquire suffi-
cient knowledge for applications of these noncoding RNAs in
clinical care.

References

1. Derrien T, Johnson R, Bussotti G, Tanzer 12. Laulagnier K, Motta C, Hamdi S, Roy S,


A, Djebali S et al (2012) The GENCODE Fauvelle F et al (2004) Mast cell- and den-
v7 catalog of human long noncoding RNAs: dritic cell-derived exosomes display a specific
analysis of their gene structure, evolution, and lipid composition and an unusual membrane
expression. Genome Res 22:1775–1789 organization. Biochem J 380:161–171
2. Bartel DP (2004) MicroRNAs: genomics, 13. Hogan MC, Manganelli L, Woollard
biogenesis, mechanism, and function. Cell JR, Masyuk AI, Masyuk TV et al (2009)
116:281–297 Characterization of PKD protein-positive
3. Lee RC, Feinbaum RL, Ambros V (1993) exosome-like vesicles. J Am Soc Nephrol
The C. elegans heterochronic gene lin-4 20:278–288
encodes small RNAs with antisense comple- 14. Zhou R, O'Hara SP, Chen XM (2011)
mentarity to lin-14. Cell 75:843–854 MicroRNA regulation of innate immune
4. Kozomara A, Griffiths-Jones S (2014) miR- responses in epithelial cells. Cell Mol
Base: annotating high confidence microRNAs Immunol 8:371–379
using deep sequencing data. Nucleic Acids 15. Vlassov AV, Magdaleno S, Setterquist
Res 42:D68–D73 R, Conrad R (2012) Exosomes: cur-
5. Lee Y, Kim M, Han J, Yeom KH, Lee S et al rent knowledge of their composition,
(2004) MicroRNA genes are transcribed by biological functions, and diagnostic and
RNA polymerase II. EMBO J 23:4051–4060 therapeutic potentials. Biochim Biophys Acta
6. Bortolin-Cavaille ML, Dance M, Weber M, 1820:940–948
Cavaille J (2009) C19MC microRNAs are 16. Mittelbrunn M, Gutierrez-Vazquez C,
processed from introns of large Pol-II, non-­ Villarroya-Beltri C, Gonzalez S, Sanchez-­
protein-­coding transcripts. Nucleic Acids Res Cabo F et al (2011) Unidirectional transfer of
37:3464–3473 microRNA-loaded exosomes from T cells to
7. Katahira J, Yoneda Y (2011) Nucleocytoplasmic antigen-presenting cells. Nat Commun 2:282
transport of microRNAs and related small 17. McDonald MK, Tian Y, Qureshi RA, Gormley
RNAs. Traffic 12:1468–1474 M, Ertel A et al (2014) Functional ­significance
8. Bhayani MK, Calin GA, Lai SY (2012) of macrophage-derived exosomes in inflam-
Functional relevance of miRNA sequences in mation and pain. Pain 155:1527–1539
human disease. Mutat Res 731:14–19 18. Putz U, Howitt J, Doan A, Goh CP, Low LH
9. Roberts TC (2014) The MicroRNA biology et al (2012) The tumor suppressor PTEN is
of the mammalian nucleus. Mol Ther Nucleic exported in exosomes and has phosphatase
Acids 3:e188 activity in recipient cells. Sci Signal 5:ra70
10. Hu G, Drescher KM, Chen XM (2012) 19. Bracken CP, Gregory PA, Kolesnikoff N,
Exosomal miRNAs: biological properties and Bert AG, Wang J et al (2008) A double-
therapeutic potential. Front Genet 3:56 negative feedback loop between ZEB1-SIP1
and the microRNA-200 family regulates
11. Valadi H, Ekstrom K, Bossios A, Sjostrand epithelial-­mesenchymal transition. Cancer
M, Lee JJ et al (2007) Exosome-mediated Res 68:7846–7854
transfer of mRNAs and microRNAs is a novel
mechanism of genetic exchange between 20. Li C, Nguyen HT, Zhuang Y, Lin
cells. Nat Cell Biol 9:654–659 Y, Flemington EK et al (2011) Post-­
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 17

transcriptional up-regulation of miR-21 by gation and chemoresistance. J Biol Chem


type I collagen. Mol Carcinog 50:563–570 288:4334–4345
21. Olsen PH, Ambros V (1999) The lin-4 regu- 34. Zhou M, Liu Z, Zhao Y, Ding Y, Liu H
latory RNA controls developmental timing in et al (2010) MicroRNA-125b confers the
Caenorhabditis elegans by blocking LIN-14 resistance of breast cancer cells to paclitaxel
protein synthesis after the initiation of trans- through suppression of pro-apoptotic Bcl-2
lation. Dev Biol 216:671–680 antagonist killer 1 (Bak1) expression. J Biol
22. Friedman RC, Farh KK, Burge CB, Bartel DP Chem 285:21496–21507
(2009) Most mammalian mRNAs are con- 35. Kiriakidou M, Tan GS, Lamprinaki S, De
served targets of microRNAs. Genome Res Planell-Saguer M, Nelson PT et al (2007) An
19:92–105 mRNA m7G cap binding-like motif within
23. Agarwal V, Bell GW, Nam JW, Bartel DP human Ago2 represses translation. Cell
(2015) Predicting effective microRNA target 129:1141–1151
sites in mammalian mRNAs. eLife 4:e05005 36. Stefani G, Slack FJ (2008) Small non-coding
24. Maroney PA, Yu Y, Fisher J, Nilsen TW (2006) RNAs in animal development. Nat Rev Mol
Evidence that microRNAs are associated with Cell Biol 9:219–230
translating messenger RNAs in human cells. 37. Kim DH, Saetrom P, Snove O Jr, Rossi JJ
Nat Struct Mol Biol 13:1102–1107 (2008) MicroRNA-directed transcriptional
25. Nottrott S, Simard MJ, Richter JD (2006) gene silencing in mammalian cells. Proc Natl
Human let-7a miRNA blocks protein pro- Acad Sci U S A 105:16230–16235
duction on actively translating polyribosomes. 38. Benhamed M, Herbig U, Ye T, Dejean A,
Nat Struct Mol Biol 13:1108–1114 Bischof O (2012) Senescence is an endog-
26. Petersen CP, Bordeleau ME, Pelletier J, Sharp enous trigger for microRNA-directed tran-
PA (2006) Short RNAs repress translation scriptional gene silencing in human cells. Nat
after initiation in mammalian cells. Mol Cell Cell Biol 14:266–275
21:533–542 39. Zardo G, Ciolfi A, Vian L, Starnes LM, Billi M
27. Pillai RS, Bhattacharyya SN, Artus CG, Zoller et al (2012) Polycombs and microRNA-223
T, Cougot N et al (2005) Inhibition of trans- regulate human granulopoiesis by transcrip-
lational initiation by Let-7 MicroRNA in tional control of target gene expression.
human cells. Science 309:1573–1576 Blood 119:4034–4046
28. Humphreys DT, Westman BJ, Martin DI, 40. Adilakshmi T, Sudol I, Tapinos N (2012)
Preiss T (2005) MicroRNAs control transla- Combinatorial action of miRNAs regulates
tion initiation by inhibiting eukaryotic initia- transcriptional and post-transcriptional gene
tion factor 4E/cap and poly(A) tail function. silencing following in vivo PNS injury. PLoS
Proc Natl Acad Sci U S A 102:16961–16966 One 7:e39674
29. Rehwinkel J, Behm-Ansmant I, Gatfield D, 41. Place RF, Li LC, Pookot D, Noonan EJ,
Izaurralde E (2005) A crucial role for GW182 Dahiya R (2008) MicroRNA-373 induces
and the DCP1:DCP2 decapping complex expression of genes with complementary pro-
in miRNA-mediated gene silencing. RNA moter sequences. Proc Natl Acad Sci U S A
11:1640–1647 105:1608–1613
30. Behm-Ansmant I, Rehwinkel J, Doerks 42. Shimakami T, Yamane D, Jangra RK, Kempf
T, Stark A, Bork P et al (2006) mRNA BJ, Spaniel C et al (2012) Stabilization of hep-
degradation by miRNAs and GW182 atitis C virus RNA by an Ago2-miR-122 com-
requires both CCR4:NOT deadenylase and plex. Proc Natl Acad Sci U S A 109:941–946
DCP1:DCP2 decapping complexes. Genes 43. Ha TY (2011) The role of MicroRNAs in reg-
Dev 20:1885–1898 ulatory T cells and in the immune response.
31. Wu L, Fan J, Belasco JG (2006) MicroRNAs Immune Netw 11:11–41
direct rapid deadenylation of mRNA. Proc 44. Calin GA, Dumitru CD, Shimizu M, Bichi
Natl Acad Sci U S A 103:4034–4039 R, Zupo S et al (2002) Frequent deletions
32. Guo H, Ingolia NT, Weissman JS, Bartel DP and down-regulation of micro-RNA genes
(2010) Mammalian microRNAs predomi- miR15 and miR16 at 13q14 in chronic lym-
nantly act to decrease target mRNA levels. phocytic leukemia. Proc Natl Acad Sci U S A
Nature 466:835–840 99:15524–15529
33. Liu Z, Liu H, Desai S, Schmitt DC, Zhou 45. Hanahan D, Weinberg RA (2011) Hallmarks
M et al (2013) miR-125b functions as a key of cancer: the next generation. Cell
mediator for snail-induced stem cell propa- 144:646–674
18 Min Xue et al.

46. He L, Thomson JM, Hemann MT, 57. Wurdinger T, Tannous BA, Saydam O, Skog
Hernando-Monge E, Mu D et al (2005) A J, Grau S et al (2008) miR-296 regulates
microRNA polycistron as a potential human growth factor receptor overexpression in
oncogene. Nature 435:828–833 angiogenic endothelial cells. Cancer Cell
47. Hayashita Y, Osada H, Tatematsu Y, Yamada 14:382–393
H, Yanagisawa K et al (2005) A polycistronic 58. Lal A, Pan Y, Navarro F, Dykxhoorn DM,
microRNA cluster, miR-17-92, is overex- Moreau L et al (2009) miR-24-mediated
pressed in human lung cancers and enhances downregulation of H2AX suppresses DNA
cell proliferation. Cancer Res 65:9628–9632 repair in terminally differentiated blood cells.
48. Takamizawa J, Konishi H, Yanagisawa K, Nat Struct Mol Biol 16:492–498
Tomida S, Osada H et al (2004) Reduced 59. Calin GA, Liu CG, Sevignani C, Ferracin M,
expression of the let-7 microRNAs in human Felli N et al (2004) MicroRNA profiling
lung cancers in association with shortened reveals distinct signatures in B cell chronic
postoperative survival. Cancer Res lymphocytic leukemias. Proc Natl Acad Sci U
64:3753–3756 S A 101:11755–11760
49. Johnson SM, Grosshans H, Shingara J, Byrom 60. Chan JA, Krichevsky AM, Kosik KS (2005)
M, Jarvis R et al (2005) RAS is regulated by MicroRNA-21 is an antiapoptotic factor in
the let-7 microRNA family. Cell human glioblastoma cells. Cancer Res
120:635–647 65:6029–6033
50. Tsang WP, Ng EK, Ng SS, Jin H, Yu J et al 61. Doghman M, El Wakil A, Cardinaud B,
(2010) Oncofetal H19-derived miR-675 Thomas E, Wang J et al (2010) Regulation of
regulates tumor suppressor RB in human insulin-like growth factor-mammalian target
colorectal cancer. Carcinogenesis 31:350– of rapamycin signaling by microRNA in child-
358 hood adrenocortical tumors. Cancer Res
51. Ueda R, Kohanbash G, Sasaki K, Fujita M, 70:4666–4675
Zhu X et al (2009) Dicer-regulated microR- 62. Ma L, Teruya-Feldstein J, Weinberg RA
NAs 222 and 339 promote resistance of can- (2007) Tumour invasion and metastasis initi-
cer cells to cytotoxic T-lymphocytes by ated by microRNA-10b in breast cancer.
down-regulation of ICAM-1. Proc Natl Acad Nature 449:682–688
Sci U S A 106:10746–10751 63. Calin GA, Sevignani C, Dumitru CD, Hyslop
52. Tazawa H, Tsuchiya N, Izumiya M, Nakagama T, Noch E et al (2004) Human microRNA
H (2007) Tumor-suppressive miR-34a genes are frequently located at fragile sites
induces senescence-like growth arrest through and genomic regions involved in cancers.
modulation of the E2F pathway in human Proc Natl Acad Sci U S A 101:2999–3004
colon cancer cells. Proc Natl Acad Sci U S A 64. Jazdzewski K, Murray EL, Franssila K, Jarzab
104:15472–15477 B, Schoenberg DR et al (2008) Common
53. O'Connell RM, Taganov KD, Boldin MP, SNP in pre-miR-146a decreases mature miR
Cheng G, Baltimore D (2007) MicroRNA-155 expression and predisposes to papillary thy-
is induced during the macrophage inflamma- roid carcinoma. Proc Natl Acad Sci U S A
tory response. Proc Natl Acad Sci U S A 105:7269–7274
104:1604–1609 65. Lee YS, Dutta A (2007) The tumor suppres-
54. Gironella M, Seux M, Xie MJ, Cano C, sor microRNA let-7 represses the HMGA2
Tomasini R et al (2007) Tumor protein oncogene. Genes Dev 21:1025–1030
53-induced nuclear protein 1 expression is 66. Mayr C, Hemann MT, Bartel DP (2007)
repressed by miR-155, and its restoration Disrupting the pairing between let-7 and
inhibits pancreatic tumor development. Proc Hmga2 enhances oncogenic transformation.
Natl Acad Sci U S A 104:16170–16175 Science 315:1576–1579
55. Gregory PA, Bert AG, Paterson EL, Barry 67. Chin LJ, Ratner E, Leng S, Zhai R, Nallur S
SC, Tsykin A et al (2008) The miR-200 fam- et al (2008) A SNP in a let-7 microRNA
ily and miR-205 regulate epithelial to mesen- ­complementary site in the KRAS 3′ untrans-
chymal transition by targeting ZEB1 and lated region increases non-small cell lung can-
SIP1. Nat Cell Biol 10:593–601 cer risk. Cancer Res 68:8535–8540
56. Nguyen HT, Li C, Lin Z, Zhuang Y, 68. Volinia S, Calin GA, Liu CG, Ambs S,
Flemington EK et al (2012) The microRNA Cimmino A et al (2006) A microRNA expres-
expression associated with morphogenesis of sion signature of human solid tumors defines
breast cancer cells in three-dimensional cancer gene targets. Proc Natl Acad Sci U S A
organotypic culture. Oncol Rep 28:117–126 103:2257–2261
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 19

69. O'Donnell KA, Wentzel EA, Zeller KI, Dang 82. Lowery AJ, Miller N, Devaney A, McNeill
CV, Mendell JT (2005) c-Myc-regulated RE, Davoren PA et al (2009) MicroRNA sig-
microRNAs modulate E2F1 expression. natures predict oestrogen receptor, progester-
Nature 435:839–843 one receptor and HER2/neu receptor status
70. Peter ME (2009) Let-7 and miR-200 microR- in breast cancer. Breast Cancer Res 11:R27
NAs: guardians against pluripotency and can- 83. Volinia S, Galasso M, Sana ME, Wise TF,
cer progression. Cell Cycle 8:843–852 Palatini J et al (2012) Breast cancer signatures
71. Li C, Nguyen HT, Zhuang Y, Lin Z, for invasiveness and prognosis defined by
Flemington EK et al (2012) Comparative deep sequencing of microRNA. Proc Natl
profiling of miRNA expression of lung adeno- Acad Sci U S A 109:3024–3029
carcinoma cells in two-dimensional and three-­ 84. Foekens JA, Sieuwerts AM, Smid M, Look
dimensional cultures. Gene 511:143–150 MP, de Weerd V et al (2008) Four miRNAs
72. Mouw JK, Yui Y, Damiano L, Bainer RO, associated with aggressiveness of lymph node-­
Lakins JN et al (2014) Tissue mechanics negative, estrogen receptor-positive human
modulate microRNA-dependent PTEN breast cancer. Proc Natl Acad Sci U S A
expression to regulate malignant progression. 105:13021–13026
Nat Med 20:360–367 85. Rodriguez-Gonzalez FG, Sieuwerts AM,
73. Weber JA, Baxter DH, Zhang S, Huang DY, Smid M, Look MP, Meijer-van Gelder ME
Huang KH et al (2010) The microRNA spec- et al (2011) MicroRNA-30c expression level
trum in 12 body fluids. Clin Chem is an independent predictor of clinical benefit
56:1733–1741 of endocrine therapy in advanced estrogen
74. Taylor DD, Gercel-Taylor C (2008) receptor positive breast cancer. Breast Cancer
MicroRNA signatures of tumor-derived exo- Res Treat 127:43–51
somes as diagnostic biomarkers of ovarian 86. Maillot G, Lacroix-Triki M, Pierredon S,
cancer. Gynecol Oncol 110:13–21 Gratadou L, Schmidt S et al (2009)
75. Skog J, Wurdinger T, van Rijn S, Meijer DH, Widespread estrogen-dependent repression of
Gainche L et al (2008) Glioblastoma microves- microRNAs involved in breast tumor cell
icles transport RNA and proteins that pro- growth. Cancer Res 69:8332–8340
mote tumour growth and provide diagnostic 87. Ichikawa T, Sato F, Terasawa K, Tsuchiya S,
biomarkers. Nat Cell Biol 10:1470–1476 Toi M et al (2012) Trastuzumab produces
76. Ng EK, Chong WW, Jin H, Lam EK, Shin VY therapeutic actions by upregulating miR-26a
et al (2009) Differential expression of microR- and miR-30b in breast cancer cells. PLoS One
NAs in plasma of patients with colorectal can- 7:e31422
cer: a potential marker for colorectal cancer 88. Ebert MS, Neilson JR, Sharp PA (2007)
screening. Gut 58:1375–1381 MicroRNA sponges: competitive inhibitors of
77. Huang Z, Huang D, Ni S, Peng Z, Sheng W small RNAs in mammalian cells. Nat Methods
et al (2010) Plasma microRNAs are promis- 4:721–726
ing novel biomarkers for early detection of 89. Ma L, Young J, Prabhala H, Pan E, Mestdagh
colorectal cancer. Int J Cancer 127:118–126 P et al (2010) miR-9, a MYC/MYCN-­
78. Pu XX, Huang GL, Guo HQ, Guo CC, Li H activated microRNA, regulates E-cadherin
et al (2010) Circulating miR-221 directly and cancer metastasis. Nat Cell Biol
amplified from plasma is a potential diagnos- 12:247–256
tic and prognostic marker of colorectal cancer 90. Huang S, Chen Y, Wu W, Ouyang N, Chen
and is correlated with p53 expression. J et al (2013) miR-150 promotes human
J Gastroenterol Hepatol 25:1674–1680 breast cancer growth and malignant behavior
79. Cheng H, Zhang L, Cogdell DE, Zheng H, by targeting the pro-apoptotic purinergic
Schetter AJ et al (2011) Circulating plasma P2X7 receptor. PLoS One 8:e80707
MiR-141 is a novel biomarker for metastatic 91. Kuss AW, Chen W (2008) MicroRNAs in
colon cancer and predicts poor prognosis. brain function and disease. Curr Neurol
PLoS One 6:e17745 Neurosci Rep 8:190–197
80. Toiyama Y, Takahashi M, Hur K, Nagasaka T, 92. Kim J, Inoue K, Ishii J, Vanti WB, Voronov
Tanaka K et al (2013) Serum miR-21 as a diag- SV et al (2007) A MicroRNA feedback circuit
nostic and prognostic biomarker in colorectal in midbrain dopamine neurons. Science
cancer. J Natl Cancer Inst 105:849–859 317:1220–1224
81. Ogata-Kawata H, Izumiya M, Kurioka D, 93. Gehrke S, Imai Y, Sokol N, Lu B (2010)
Honma Y, Yamada Y et al (2014) Circulating Pathogenic LRRK2 negatively regulates
exosomal microRNAs as biomarkers of colon microRNA-mediated translational repression.
cancer. PLoS One 9:e92921 Nature 466:637–641
20 Min Xue et al.

94. Blennow K, de Leon MJ, Zetterberg H 106. Jardim MJ, Dailey L, Silbajoris R, Diaz-­
(2006) Alzheimer’s disease. Lancet Sanchez D (2012) Distinct microRNA
368:387–403 expression in human airway cells of asthmatic
95. Cogswell JP, Ward J, Taylor IA, Waters M, donors identifies a novel asthma-associated
Shi Y et al (2008) Identification of miRNA gene. Am J Respir Cell Mol Biol
changes in Alzheimer's disease brain and CSF 47:536–542
yields putative biomarkers and insights into 107. Solberg OD, Ostrin EJ, Love MI, Peng JC,
disease pathways. J Alzheimers Dis 14:27–41 Bhakta NR et al (2012) Airway epithelial
96. Maes OC, Chertkow HM, Wang E, Schipper miRNA expression is altered in asthma. Am
HM (2009) MicroRNA: implications for J Respir Crit Care Med 186:965–974
Alzheimer disease and other human CNS dis- 108. Tsitsiou E, Williams AE, Moschos SA, Patel
orders. Curr Genomics 10:154–168 K, Rossios C et al (2012) Transcriptome anal-
97. Maes OC, Xu S, Yu B, Chertkow HM, Wang ysis shows activation of circulating CD8+ T
E et al (2007) Transcriptional profiling of cells in patients with severe asthma. J Allergy
Alzheimer blood mononuclear cells by micro- Clin Immunol 129:95–103
array. Neurobiol Aging 28:1795–1809 109. O'Connell RM, Rao DS, Baltimore D (2012)
98. Blalock EM, Chen KC, Stromberg AJ, Norris microRNA regulation of inflammatory
CM, Kadish I et al (2005) Harnessing the responses. Annu Rev Immunol 30:295–312
power of gene microarrays for the study of 110. Pandit KV, Corcoran D, Yousef H, Yarlagadda
brain aging and Alzheimer’s disease: statistical M, Tzouvelekis A et al (2010) Inhibition and
reliability and functional correlation. Ageing role of let-7d in idiopathic pulmonary fibro-
Res Rev 4:481–512 sis. Am J Respir Crit Care Med
99. Nunez-Iglesias J, Liu CC, Morgan TE, Finch 182:220–229
CE, Zhou XJ (2010) Joint genome-wide pro- 111. Dakhlallah D, Batte K, Wang Y, Cantemir-­
filing of miRNA and mRNA expression in Stone CZ, Yan P et al (2013) Epigenetic reg-
Alzheimer's disease cortex reveals altered ulation of miR-17~92 contributes to the
miRNA regulation. PLoS One 5:e8898 pathogenesis of pulmonary fibrosis. Am
100. Hebert SS, Horre K, Nicolai L, Papadopoulou J Respir Crit Care Med 187:397–405
AS, Mandemakers W et al (2008) Loss of 112. Liu G, Friggeri A, Yang Y, Milosevic J, Ding
microRNA cluster miR-29a/b-1 in sporadic Q et al (2010) miR-21 mediates fibrogenic
Alzheimer’s disease correlates with increased activation of pulmonary fibroblasts and lung
BACE1/beta-secretase expression. Proc Natl fibrosis. J Exp Med 207:1589–1597
Acad Sci U S A 105:6415–6420 113. Cushing L, Kuang PP, Qian J, Shao F, Wu
101. Hebert SS, Horre K, Nicolai L, Bergmans B, J et al (2011) miR-29 is a major regulator of
Papadopoulou AS et al (2009) MicroRNA genes associated with pulmonary fibrosis. Am
regulation of Alzheimer’s Amyloid precursor J Respir Cell Mol Biol 45:287–294
protein expression. Neurobiol Dis 114. Yang S, Banerjee S, de Freitas A, Sanders YY,
33:422–428 Ding Q et al (2012) Participation of miR-­
102. Booton R, Lindsay MA (2014) Emerging 200 in pulmonary fibrosis. Am J Pathol
role of MicroRNAs and long noncoding 180:484–493
RNAs in respiratory disease. Chest 115. Jiang X, Tsitsiou E, Herrick SE, Lindsay MA
146:193–204 (2010) MicroRNAs and the regulation of
103. Williams AE, Moschos SA, Perry MM, Barnes fibrosis. FEBS J 277:2015–2021
PJ, Lindsay MA (2007) Maternally imprinted 116. Ezzie ME, Crawford M, Cho JH, Orellana R,
microRNAs are differentially expressed dur- Zhang S et al (2012) Gene expression net-
ing mouse and human lung development. works in COPD: microRNA and mRNA reg-
Dev Dyn 236:572–580 ulation. Thorax 67:122–131
104. Harris KS, Zhang Z, McManus MT, Harfe 117. Sato T, Liu X, Nelson A, Nakanishi M, Kanaji
BD, Sun X (2006) Dicer function is essential N et al (2010) Reduced miR-146a increases
for lung epithelium morphogenesis. Proc prostaglandin E(2)in chronic obstructive pul-
Natl Acad Sci U S A 103:2208–2213 monary disease fibroblasts. Am J Respir Crit
105. Lu Y, Thomson JM, Wong HY, Hammond Care Med 182:1020–1029
SM, Hogan BL (2007) Transgenic over-­ 118. Lewis A, Riddoch-Contreras J, Natanek SA,
expression of the microRNA miR-17-92 clus- Donaldson A, Man WD et al (2012)
ter promotes proliferation and inhibits Downregulation of the serum response fac-
differentiation of lung epithelial progenitor tor/miR-1 axis in the quadriceps of patients
cells. Dev Biol 310:442–453 with COPD. Thorax 67:26–34
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 21

119. Cunningham F, Amode MR, Barrell D, Beal RNA mediates transcriptional gene silencing
K, Billis K et al (2015) Ensembl 2015. Nucleic by interacting with Dnmt1. Development
Acids Res 43:D662–D669 137:2493–2499
120. Lanz RB, McKenna NJ, Onate SA, Albrecht 133. Wu Y, Zhang L, Wang Y, Li H, Ren X et al
U, Wong J et al (1999) A steroid receptor (2015) Long non-coding RNA HOTAIR
coactivator, SRA, functions as an RNA and is promotes tumor cell invasion and metastasis
present in an SRC-1 complex. Cell 97:17–27 by recruiting EZH2 and repressing E-cadherin
121. Rinn JL, Chang HY (2012) Genome regula- in oral squamous cell carcinoma. Int J Oncol
tion by long noncoding RNAs. Annu Rev 46:2586–2594
Biochem 81:145–166 134. Khalil AM, Guttman M, Huarte M, Garber
122. Shibayama Y, Fanucchi S, Magagula L, M, Raj A et al (2009) Many human large
Mhlanga MM (2014) lncRNA and gene intergenic noncoding RNAs associate with
looping: what’s the connection? Transcription chromatin-modifying complexes and affect
5:e28658 gene expression. Proc Natl Acad Sci U S A
123. Shi X, Sun M, Liu H, Yao Y, Song Y (2013) 106:11667–11672
Long non-coding RNAs: a new frontier in the 135. Zhao J, Ohsumi TK, Kung JT, Ogawa Y,
study of human diseases. Cancer Lett Grau DJ et al (2010) Genome-wide identifi-
339:159–166 cation of polycomb-associated RNAs by RIP-­
124. Zong X, Tripathi V, Prasanth KV (2011) seq. Mol Cell 40:939–953
RNA splicing control: yet another gene regu- 136. Brannan CI, Dees EC, Ingram RS, Tilghman
latory role for long nuclear noncoding RNAs. SM (1990) The product of the H19 gene
RNA Biol 8:968–977 may function as an RNA. Mol Cell Biol
125. Kretz M, Siprashvili Z, Chu C, Webster DE, 10:28–36
Zehnder A et al (2013) Control of somatic 137. Brown CJ, Ballabio A, Rupert JL, Lafreniere
tissue differentiation by the long non-coding RG, Grompe M et al (1991) A gene from the
RNA TINCR. Nature 493:231–235 region of the human X inactivation centre is
126. Hacisuleyman E, Goff LA, Trapnell C, expressed exclusively from the inactive X
Williams A, Henao-Mejia J et al (2014) chromosome. Nature 349:38–44
Topological organization of multichromo- 138. Simon MD, Pinter SF, Fang R, Sarma K,
somal regions by the long intergenic noncod- Rutenberg-Schoenberg M et al (2013) High-­
ing RNA Firre. Nat Struct Mol Biol resolution Xist binding maps reveal two-step
21:198–206 spreading during X-chromosome inactiva-
127. Ponting CP, Oliver PL, Reik W (2009) tion. Nature 504:465–469
Evolution and functions of long noncoding 139. Chu C, Qu K, Zhong FL, Artandi SE, Chang
RNAs. Cell 136:629–641 HY (2011) Genomic maps of long noncoding
128. Nagano T, Mitchell JA, Sanz LA, Pauler FM, RNA occupancy reveal principles of RNA-­
Ferguson-Smith AC et al (2008) The Air chromatin interactions. Mol Cell
noncoding RNA epigenetically silences tran- 44:667–678
scription by targeting G9a to chromatin. 140. Arab K, Park YJ, Lindroth AM, Schafer A,
Science 322:1717–1720 Oakes C et al (2014) Long noncoding RNA
129. Rinn JL, Kertesz M, Wang JK, Squazzo SL, TARID directs demethylation and activation
Xu X et al (2007) Functional demarcation of of the tumor suppressor TCF21 via
active and silent chromatin domains in human GADD45A. Mol Cell 55(4):604–614
HOX loci by noncoding RNAs. Cell 141. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q
129:1311–1323 et al (2010) The nuclear-retained noncoding
130. Pandey RR, Mondal T, Mohammad F, Enroth RNA MALAT1 regulates alternative splicing
S, Redrup L et al (2008) Kcnq1ot1 antisense by modulating SR splicing factor phosphory-
noncoding RNA mediates lineage-specific lation. Mol Cell 39:925–938
transcriptional silencing through chromatin-­ 142. Bernard D, Prasanth KV, Tripathi V, Colasse
level regulation. Mol Cell 32:232–246 S, Nakamura T et al (2010) A long nuclear-­
131. Zhao J, Sun BK, Erwin JA, Song JJ, Lee JT retained non-coding RNA regulates synapto-
(2008) Polycomb proteins targeted by a short genesis by modulating gene expression.
repeat RNA to the mouse X chromosome. EMBO J 29:3082–3093
Science 322:750–756 143. Gong C, Maquat LE (2011) lncRNAs trans-
132. Mohammad F, Mondal T, Guseva N, Pandey activate STAU1-mediated mRNA decay by
GK, Kanduri C (2010) Kcnq1ot1 noncoding duplexing with 3′ UTRs via Alu elements.
Nature 470:284–288
22 Min Xue et al.

144. Yoon JH, Abdelmohsen K, Srikantan S, Yang 156. Naemura M, Murasaki C, Inoue Y, Okamoto
X, Martindale JL et al (2012) LincRNA-p21 H, Kotake Y (2015) Long noncoding RNA
suppresses target mRNA translation. Mol Cell ANRIL regulates proliferation of non-small
47:648–655 cell lung cancer and cervical cancer cells.
145. Carrieri C, Cimatti L, Biagioli M, Beugnet A, Anticancer Res 35:5377–5382
Zucchelli S et al (2012) Long non-coding 157. Cai Y, He J, Zhang D (2015) Long noncod-
antisense RNA controls Uchl1 translation ing RNA CCAT2 promotes breast tumor
through an embedded SINEB2 repeat. growth by regulating the Wnt signaling path-
Nature 491:454–457 way. Onco Targets Ther 8:2657–2664
146. Zappulla DC, Cech TR (2006) RNA as a flex- 158. Zhuang Y, Nguyen HT, Burow ME, Zhuo Y,
ible scaffold for proteins: yeast telomerase and El-Dahr SS et al (2014) Elevated expression
beyond. Cold Spring Harb Symp Quant Biol of long intergenic non-coding RNA HOTAIR
71:217–224 in a basal-like variant of MCF-7 breast cancer
147. Koziol MJ, Rinn JL (2010) RNA traffic con- cells. Mol Carcinog. 54(12):1656–1667
trol of chromatin complexes. Curr Opin 159. Gupta RA, Shah N, Wang KC, Kim J,
Genet Dev 20:142–148 Horlings HM et al (2010) Long non-coding
148. Tsai MC, Manor O, Wan Y, Mosammaparast RNA HOTAIR reprograms chromatin state
N, Wang JK et al (2010) Long noncoding to promote cancer metastasis. Nature
RNA as modular scaffold of histone modifica- 464:1071–1076
tion complexes. Science 329:689–693 160. Kogo R, Shimamura T, Mimori K, Kawahara
149. Faghihi MA, Modarresi F, Khalil AM, Wood K, Imoto S et al (2011) Long noncoding
DE, Sahagan BG et al (2008) Expression of a RNA HOTAIR regulates polycomb-­
noncoding RNA is elevated in Alzheimer’s dependent chromatin modification and is
disease and drives rapid feed-forward regula- associated with poor prognosis in colorectal
tion of beta-secretase. Nat Med 14:723–730 cancers. Cancer Res 71:6320–6326
150. Carrieri C, Forrest AR, Santoro C, Persichetti 161. Chung S, Nakagawa H, Uemura M, Piao L,
F, Carninci P et al (2015) Expression analysis Ashikawa K et al (2011) Association of a
of the long non-coding RNA antisense to novel long non-coding RNA in 8q24 with
Uchl1 (AS Uchl1) during dopaminergic cells’ prostate cancer susceptibility. Cancer Sci
differentiation in vitro and in neurochemical 102:245–252
models of Parkinson’s disease. Front Cell 162. Calin GA, Pekarsky Y, Croce CM (2007) The
Neurosci 9:114 role of microRNA and other non-coding
151. Ishii N, Ozaki K, Sato H, Mizuno H, Saito S RNA in the pathogenesis of chronic lympho-
et al (2006) Identification of a novel non-­ cytic leukemia. Best Pract Res Clin Haematol
coding RNA, MIAT, that confers risk of myo- 20:425–437
cardial infarction. J Hum Genet 163. Calin GA, Liu CG, Ferracin M, Hyslop T,
51:1087–1099 Spizzo R et al (2007) Ultraconserved regions
152. Pasmant E, Laurendeau I, Heron D, Vidaud encoding ncRNAs are altered in human leuke-
M, Vidaud D et al (2007) Characterization of mias and carcinomas. Cancer Cell 12:215–229
a germ-line deletion, including the entire 164. Khaitan D, Dinger ME, Mazar J, Crawford J,
INK4/ARF locus, in a melanoma-neural sys- Smith MA et al (2011) The melanoma-­
tem tumor family: identification of ANRIL, upregulated long noncoding RNA
an antisense noncoding RNA whose expres- SPRY4-IT1 modulates apoptosis and inva-
sion coclusters with ARF. Cancer Res sion. Cancer Res 71:3852–3862
67:3963–3969 165. Li L, Feng T, Lian Y, Zhang G, Garen A et al
153. Daughters RS, Tuttle DL, Gao W, Ikeda Y, (2009) Role of human noncoding RNAs in
Moseley ML et al (2009) RNA gain-of-­ the control of tumorigenesis. Proc Natl Acad
function in spinocerebellar ataxia type 8. Sci U S A 106:12956–12961
PLoS Genet 5:e1000600 166. Huarte M, Guttman M, Feldser D, Garber
154. Khalil AM, Faghihi MA, Modarresi F, M, Koziol MJ et al (2010) A large intergenic
Brothers SP, Wahlestedt C (2008) A novel noncoding RNA induced by p53 mediates
RNA transcript with antiapoptotic function is global gene repression in the p53 response.
silenced in fragile X syndrome. PLoS One Cell 142:409–419
3:e1486 167. Yu W, Gius D, Onyango P, Muldoon-Jacobs
155. Prensner JR, Chinnaiyan AM (2011) The K, Karp J et al (2008) Epigenetic silencing of
emergence of lncRNAs in cancer biology. tumour suppressor gene p15 by its antisense
Cancer Discov 1:391–407 RNA. Nature 451:202–206
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 23

168. Gutschner T, Hammerle M, Eissmann M, 179. Wang X, Li M, Wang Z, Han S, Tang X et al


Hsu J, Kim Y et al (2013) The noncoding (2015) Silencing of long noncoding RNA
RNA MALAT1 is a critical regulator of the MALAT1 by miR-101 and miR-217 inhibits
metastasis phenotype of lung cancer cells. proliferation, migration, and invasion of
Cancer Res 73:1180–1189 esophageal squamous cell carcinoma cells.
169. Tano K, Mizuno R, Okada T, Rakwal R, J Biol Chem 290:3925–3935
Shibato J et al (2010) MALAT-1 enhances 180. Mohamadkhani A (2014) Long noncoding
cell motility of lung adenocarcinoma cells by RNAs in interaction with RNA binding pro-
influencing the expression of motility-related teins in hepatocellular carcinoma. Hepat Mon
genes. FEBS Lett 584:4575–4580 14:e18794
170. Lopez-Ayllon BD, Moncho-Amor V, 181. Liu SP, Yang JX, Cao DY, Shen K (2013)
Abarrategi A, Ibanez de Caceres I, Castro-­ Identification of differentially expressed long
Carpeno J et al (2014) Cancer stem cells and non-coding RNAs in human ovarian cancer
cisplatin-resistant cells isolated from non-­ cells with different metastatic potentials.
small-­lung cancer cell lines constitute related Cancer Biol Med 10:138–141
cell populations. Cancer Med 3:1099–1111 182. Ren S, Liu Y, Xu W, Sun Y, Lu J et al (2013)
171. Weber DG, Johnen G, Casjens S, Bryk O, Long noncoding RNA MALAT-1 is a new
Pesch B et al (2013) Evaluation of long non- potential therapeutic target for castration
coding RNA MALAT1 as a candidate blood-­ resistant prostate cancer. J Urol
based biomarker for the diagnosis of non-small 190:2278–2287
cell lung cancer. BMC Res Notes 6:518 183. Sowalsky AG, Xia Z, Wang L, Zhao H, Chen
172. Yao Y, Fan Y, Wu J, Wan H, Wang J et al S et al (2015) Whole transcriptome sequenc-
(2012) Potential application of non-small cell ing reveals extensive unspliced mRNA in met-
lung cancer-associated autoantibodies to early astatic castration-resistant prostate cancer.
cancer diagnosis. Biochem Biophys Res Mol Cancer Res 13:98–106
Commun 423:613–619 184. Wang F, Ren S, Chen R, Lu J, Shi X et al
173. Guffanti A, Iacono M, Pelucchi P, Kim N, (2014) Development and prospective multi-
Solda G et al (2009) A transcriptional sketch center evaluation of the long noncoding RNA
of a primary human breast cancer by 454 MALAT-1 as a diagnostic urinary biomarker
deep sequencing. BMC Genomics 10:163 for prostate cancer. Oncotarget
174. Kan JY, Wu DC, Yu FJ, Wu CY, Ho YW et al 5:11091–11102
(2015) Chemokine (C-C motif) ligand 5 is 185. Ellis MJ, Ding L, Shen D, Luo J, Suman VJ
involved in tumor-associated dendritic cell-­ et al (2012) Whole-genome analysis informs
mediated colon cancer progression through breast cancer response to aromatase inhibi-
non-coding RNA MALAT-1. J Cell Physiol tion. Nature 486:353–360
230:1883–1894 186. Ji Q, Zhang L, Liu X, Zhou L, Wang W et al
175. Fan Y, Shen B, Tan M, Mu X, Qin Y et al (2014) Long non-coding RNA MALAT1
(2014) TGF-beta-induced upregulation of promotes tumour growth and metastasis in
malat1 promotes bladder cancer metastasis by colorectal cancer through binding to SFPQ
associating with suz12. Clin Cancer Res and releasing oncogene PTBP2 from SFPQ/
20:1531–1541 PTBP2 complex. Br J Cancer 111:736–748
176. Okugawa Y, Toiyama Y, Hur K, Toden S, 187. Xu C, Yang M, Tian J, Wang X, Li Z (2011)
Saigusa S et al (2014) Metastasis-associated MALAT-1: a long non-coding RNA and its
long non-coding RNA drives gastric cancer important 3′ end functional motif in colorec-
development and promotes peritoneal metas- tal cancer metastasis. Int J Oncol
tasis. Carcinogenesis 35:2731–2739 39:169–175
177. Hu L, Wu Y, Tan D, Meng H, Wang K et al 188. Lin R, Roychowdhury-Saha M, Black C, Watt
(2015) Up-regulation of long noncoding AT, Marcusson EG et al (2011) Control of
RNA MALAT1 contributes to proliferation RNA processing by a large non-coding RNA
and metastasis in esophageal squamous cell over-expressed in carcinomas. FEBS Lett
carcinoma. J Exp Clin Cancer Res 34:7 585:671–676
178. Kuo IY, Wu CC, Chang JM, Huang YL, Lin 189. Yang L, Lin C, Liu W, Zhang J, Ohgi KA et al
CH et al (2014) Low SOX17 expression is a (2011) ncRNA- and Pc2 methylation-­
prognostic factor and drives transcriptional dependent gene relocation between nuclear
dysregulation and esophageal cancer progres- structures mediates gene activation programs.
sion. Int J Cancer 135:563–573 Cell 147:773–788
24 Min Xue et al.

190. Loewen G, Jayawickramarajah J, Zhuo Y, pressed in prostate cancer. Cancer Res


Shan B (2014) Functions of lncRNA 59:5975–5979
HOTAIR in lung cancer. J Hematol Oncol 202. Nilsson J, Skog J, Nordstrand A, Baranov V,
7:90 Mincheva-Nilsson L et al (2009) Prostate
191. Loewen G, Zhuo Y, Zhuang Y, cancer-derived urine exosomes: a novel
Jayawickramarajah J, Shan B (2014) lincRNA approach to biomarkers for prostate cancer.
HOTAIR as a novel promoter of cancer pro- Br J Cancer 100:1603–1607
gression. J Can Res Updates 3:7 203. Kogure T, Yan IK, Lin WL, Patel T (2013)
192. Zhuang Y, Wang X, Nguyen HT, Zhuo Y, Extracellular vesicle-mediated transfer of a
Cui X et al (2013) Induction of long inter- novel long noncoding RNA TUC339: a mech-
genic non-coding RNA HOTAIR in lung anism of intercellular signaling in human hepa-
cancer cells by type I collagen. J Hematol tocellular cancer. Genes Cancer 4:261–272
Oncol 6:35 204. Zhuang Y, Nguyen HT, Burow ME, Zhuo Y,
193. Svoboda M, Slyskova J, Schneiderova M, El-Dahr SS et al (2015) Elevated expression
Makovicky P, Bielik L et al (2014) HOTAIR of long intergenic non-coding RNA HOTAIR
long non-coding RNA is a negative prognos- in a basal-like variant of MCF-7 breast cancer
tic factor not only in primary tumors, but also cells. Mol Carcinog 54:1656–1667
in the blood of colorectal cancer patients. 205. Pedram Fatemi R, Salah-Uddin S, Modarresi F,
Carcinogenesis 35:1510–1515 Khoury N, Wahlestedt C et al (2015) Screening
194. Chiyomaru T, Yamamura S, Fukuhara S, for small-molecule modulators of long noncod-
Yoshino H, Kinoshita T et al (2013) Genistein ing RNA-protein interactions using
inhibits prostate cancer cell growth by target- AlphaScreen. J Biomol Screen 20:1132–1141
ing miR-34a and oncogenic HOTAIR. PLoS 206. Ng SY, Lin L, Soh BS, Stanton LW (2013)
One 8:e70372 Long noncoding RNAs in development and
195. Xue X, Yang YA, Zhang A, Fong KW, Kim disease of the central nervous system. Trends
J et al (2016) LncRNA HOTAIR enhances Genet 29:461–468
ER signaling and confers tamoxifen resistance 207. Ng SY, Johnson R, Stanton LW (2012)
in breast cancer. Oncogene Human long non-coding RNAs promote plu-
35(21):2746–2755 ripotency and neuronal differentiation by
196. Mourtada-Maarabouni M, Pickard MR, association with chromatin modifiers and
Hedge VL, Farzaneh F, Williams GT (2009) transcription factors. EMBO J 31:522–533
GAS5, a non-protein-coding RNA, controls 208. Modarresi F, Faghihi MA, Patel NS, Sahagan
apoptosis and is downregulated in breast can- BG, Wahlestedt C et al (2011) Knockdown of
cer. Oncogene 28:195–208 BACE1-AS Nonprotein-coding transcript
197. Dimitrova N, Zamudio JR, Jong RM, Soukup modulates beta-amyloid-related hippocampal
D, Resnick R et al (2014) LincRNA-p21 acti- neurogenesis. Int J Alzheimers Dis
vates p21 in cis to promote Polycomb target 2011:929042
gene expression and to enforce the G1/S 209. Muddashetty R, Khanam T, Kondrashov A,
checkpoint. Mol Cell 54:777–790 Bundman M, Iacoangeli A et al (2002)
198. Cai B, Wu Z, Liao K, Zhang S (2014) Long Poly(A)-binding protein is associated with
noncoding RNA HOTAIR can serve as a neuronal BC1 and BC200 ribonucleoprotein
common molecular marker for lymph node particles. J Mol Biol 321:433–445
metastasis: a meta-analysis. Tumour Biol 210. Mus E, Hof PR, Tiedge H (2007) Dendritic
35(9):8445–8450 BC200 RNA in aging and in Alzheimer’s dis-
199. Zheng HT, Shi DB, Wang YW, Li XX, Xu Y ease. Proc Natl Acad Sci U S A
et al (2014) High expression of lncRNA 104:10679–10684
MALAT1 suggests a biomarker of poor prog- 211. Morais VA, Verstreken P, Roethig A, Smet J,
nosis in colorectal cancer. Int J Clin Exp Snellinx A et al (2009) Parkinson's disease
Pathol 7:3174–3181 mutations in PINK1 result in decreased
200. de Kok JB, Verhaegh GW, Roelofs RW, Complex I activity and deficient synaptic
Hessels D, Kiemeney LA et al (2002) function. EMBO Mol Med 1:99–111
DD3(PCA3), a very sensitive and specific 212. Scheele C, Petrovic N, Faghihi MA, Lassmann
marker to detect prostate tumors. Cancer Res T, Fredriksson K et al (2007) The human
62:2695–2698 PINK1 locus is regulated in vivo by a non-­
201. Bussemakers MJ, van Bokhoven A, Verhaegh coding natural antisense RNA during modu-
GW, Smit FP, Karthaus HF et al (1999) DD3: lation of mitochondrial function. BMC
a new prostate-specific gene, highly overex- Genomics 8:74
MicroRNAs, Long Noncoding RNAs, and Their Functions in Human Disease 25

213. Herriges MJ, Swarr DT, Morley MP, Rathi 219. Liu XH, Sun M, Nie FQ, Ge YB, Zhang EB
KS, Peng T et al (2014) Long noncoding et al (2014) Lnc RNA HOTAIR functions as
RNAs are spatially correlated with transcrip- a competing endogenous RNA to regulate
tion factors and regulate lung development. HER2 expression by sponging miR-331-3p
Genes Dev 28:1363–1379 in gastric cancer. Mol Cancer 13:92
214. Szafranski P, Dharmadhikari AV, Brosens E, 220. Ma MZ, Li CX, Zhang Y, Weng MZ, Zhang
Gurha P, Kolodziejska KE et al (2013) Small MD et al (2014) Long non-coding RNA
noncoding differentially methylated copy-­ HOTAIR, a c-Myc activated driver of malig-
number variants, including lncRNA genes, nancy, negatively regulates miRNA-130a in
cause a lethal lung developmental disorder. gallbladder cancer. Mol Cancer 13:156
Genome Res 23:23–33 221. Zhou X, Ye F, Yin C, Zhuang Y, Yue G et al
215. Michalik KM, You X, Manavski Y, (2015) The interaction between MiR-141
Doddaballapur A, Zornig M et al (2014) and lncRNA-H19 in regulating cell prolifera-
Long noncoding RNA MALAT1 regulates tion and migration in gastric cancer. Cell
endothelial cell function and vessel growth. Physiol Biochem 36:1440–1452
Circ Res 114:1389–1397 222. Kallen AN, Zhou XB, Xu J, Qiao C, Ma J et al
216. Yoon JH, Abdelmohsen K, Gorospe M (2013) The imprinted H19 lncRNA antago-
(2014) Functional interactions among nizes let-7 microRNAs. Mol Cell 52:101–112
microRNAs and long noncoding RNAs. 223. Cesana M, Cacchiarelli D, Legnini I, Santini
Semin Cell Dev Biol 34C:9–14 T, Sthandier O et al (2011) A long noncod-
217. de Giorgio A, Krell J, Harding V, Stebbing J, ing RNA controls muscle differentiation by
Castellano L (2013) Emerging roles of com- functioning as a competing endogenous
peting endogenous RNAs in cancer: insights RNA. Cell 147:358–369
from the regulation of PTEN. Mol Cell Biol 224. Jeck WR, Sharpless NE (2014) Detecting and
33:3976–3982 characterizing circular RNAs. Nat Biotechnol
218. Wang Y, Xu Z, Jiang J, Xu C, Kang J et al 32:453–461
(2013) Endogenous miRNA sponge 225. Hansen TB, Jensen TI, Clausen BH, Bramsen
lincRNA-­ RoR regulates Oct4, Nanog, and JB, Finsen B et al (2013) Natural RNA circles
Sox2 in human embryonic stem cell self-­ function as efficient microRNA sponges.
renewal. Dev Cell 25:69–80 Nature 495:384–388
Chapter 2

MicroRNA Expression: Protein Participants in MicroRNA


Regulation
Valeria M. King and Glen M. Borchert

Abstract
MiRNAs are ~20 nt small RNAs that regulate networks of proteins using a seed region of nucleotides 2–8
to complement the 3′ UTR of target mRNAs. The biogenesis and function of miRNAs as translational
repressors is facilitated by protein counterparts that process primary and precursor miRNAs to maturity
(Drosha/DCGR8 and Dicer/TRBP respectively) and incorporate miRNAs into the protein complex
RISC to recognize and repress target mRNAs (RISC proteins: Ago/TRBP1/TRBP2/DICER). Similarly,
siRNAs through comparable mechanisms are loaded into the protein complex RITS to heterochromatin
formation of DNA and suppress transcription of particular genes. MiRNAs are also regulated themselves
through many different pathways including transcriptional regulation, post-transcriptional RNA editing,
and RNA tailing. Dysregulation of miRNAs and the protein participants that mature them are implicated
in the development of a number of diseases, tumorigenesis, and arrested development of embryonic cells.
In this chapter, we will explore the biosynthesis, function, and regulation of miRNAs.

Key words Dicer, Drosha, miRNA, mRNA, Protein, RISC, Regulation

1 MicroRNA Production and Activity

MicroRNAs are small noncoding RNAs around 22 nucleotides in


length that are involved in the regulation of mRNAs in the cyto-
plasm via inducing translational repression or message degradation
[1]. MiRNAs constitute a broad regulatory network with one
miRNA potentially regulating dozens of distinct mRNAs. These
regulatory networks control levels of specific proteins and other
RNAs like long noncoding RNAs (lncRNAs) in cells. MiRNA mis-
regulation is implicated in cancer, a myriad of other illnesses, and
abnormal development [2–4]. The first miRNA was described in
1993 and was initially thought to be a novel molecular species
unique to Caenorhabditis elegans [5]. In 2001, however, nearly 10
years after their initial discovery, miRNAs were found to occur in
several different species including humans [6]. Since that time,
novel miRNA discovery has proceeded at a marked rate, and by

Jingshan Huang et al. (eds.), Bioinformatics in MicroRNA Research, Methods in Molecular Biology, vol. 1617,
DOI 10.1007/978-1-4939-7046-9_2, © Springer Science+Business Media LLC 2017

27
28 Valeria M. King and Glen M. Borchert

2015, MiRBase.org [7] has detailed significant evidence support-


ing the expressions of over 2500 unique human miRNAs and well
over 28,000 unique miRNAs across species.

1.1 MiRNA MiRNAs are transcribed from the genome by RNA Polymerase II
Processing (Pol II) or Pol III [1]. Transcription results in an initial transcript
(called a primary miRNA or pri-miRNA) of variable length con-
taining an unprocessed hairpin [8]. The pri-miRNA is next pro-
cessed by a ribonuclease protein complex including DROSHA,
which targets and cleaves the flanking ends of the hairpin, and
DCGR8 that stabilizes the complex on the pri-miRNA. DROSHA
processing yields an ~70–100 nt long stem loop called a precursor
miRNA or pre-miRNA [9].
Following excision, the pre-miRNA stem loop is transported
out of the nucleus and into the cytoplasm via the transport protein
exportin-5 using an active transport mechanism with GTP. Once
in the cytoplasm, the pre-miRNA is targeted by another ribonucle-
ase, DICER, which cleaves the molecule further by removing the
loop portion of the hairpin and leaving an intermediate duplex
which consists of the mature miRNA and a semi-complementary
sequence referred to as the passenger strand. The intermediate
duplex, which is ~22 base pairs in length, is then loaded into an
Argonaute (Ago) protein, and the passenger strand discarded [1]
(see Fig. 1).

1.2 Genomic Loci MiRNAs can be separated into two broad categories depending on
their position in the genome: canonical and noncanonical [1].
Canonical miRNAs are those that are found in intergenic regions
and are cleaved by Drosha/DCGR1 to form the precursor miRNA
(pre-miRNA) [1, 2]. Noncanonical miRNAs are mitrons or pre-­
miRNAs that are cleaved from intron sequences using splicing
instead of Drosha.
While the evolutionary origin of miRNAs is still largely
unknown, significant evidence suggests that miRNAs and their
regulatory networks arose from the insertion of transposable ele-
ments in the genome [10]. Importantly, the ability of miRNAs to
regulate multiple distinct genes may have directly arisen as a conse-
quence of transposons inserting themselves into the UTRs of pro-
tein coding genes. Since miRNAs target mRNAs through sequence
complementarity, the ability of a miRNA to identify and target a
specific mRNA may well be due to a common molecular origin
shared by a miRNA locus and its mRNA target sites [10–12] (see
Fig. 2).

1.3 Translational The mature miRNA in conjunction with the Ago protein is called
Repression and Signal an RNA-induced silencing complex or RISC. MiRNAs function as
Degradation protein level regulators by binding target mRNAs and inducing
transcriptional repression and in some instances complete signal
MicroRNA Expression: Protein Participants in MicroRNA Regulation 29

Fig. 1 Canonical and noncanonical miRNA maturation pathways. Cartoon illustrating the transcription of a
canonical miRNA, Drosha/DGCR8 processing of the primary hairpin, and the export of the resulting hairpin to
the cytoplasm mediated by exportin-5 and GTP. Once in the cytoplasm, the hairpin is recognized and bound by
DICER which cleaves the loop off of the stem loop and leaves a double-stranded intermediate duplex. The
mature strand bound to the passenger strand is then loaded into an Ago protein complex called RISC. RISC
transports the miRNA to a prospective mRNA target which the miRNA recognizes and binds. Once the RISC
complex is bound to an mRNA, translation of the mRNA cannot occur and is repressed

degradation. Included in a mature miRNA is a ~7 nt sequence (nts


2–8) called the miRNA seed that perfectly complements a specific
region of a mRNA called a seed match usually found in the 3′
UTR. A miRNA seed binds a corresponding seed match in a
mRNA, and RISC inhibits its translation. RISC localizes mRNAs
that are under transcriptional repression to p-bodies where the
mRNA is eventually degraded or released back into the cytoplasm
for translation [4]. If the seed is highly complementary to the seed
match, the Ago2 protein associated in RISC cleaves the mRNA
and results in signal degradation (see Fig. 3).

1.4 RISC The regulation of mRNAs by miRNAs is accomplished by the


RNA-induced silencing complex, or RISC. RISC consists of sev-
eral different proteins, is between 200 and 500 kDa and exhibits
30 Valeria M. King and Glen M. Borchert

Fig. 2 MiRNA regulatory networks arising from transposable elements. Cartoon depicting transposable line
elements being transcribed and inserted into the genome in multiple locations including in the untranslated
region of a protein-coding gene. To produce a miRNA hairpin, two line elements are inserted in the genome
juxtaposed to each other on different strands. MiRNAs are able to recognize target mRNA seed matches
because they each share part of a line element sequence

ribonuclease activity [13]. MRNAs are incorporated into the RISC


complex when targeted by miRNAs. AGO proteins in RISC cleave
mRNAs that are highly complementary to the incorporated
miRNA, whereas mRNAs that are mostly imperfectly bound to the
miRNA are silenced by translation inhibition. The best described
protein subunits of RISC are the RNases Dicer, Ago 1 and 2. Ago
1 and 2 serve as the central components of RISC and are respon-
sible for translational repression and cleavage/degradation [14].
Also included in RISC are TAR RNA binding proteins, or TARBPs,
which comes in two isoforms, TARBP1 and 2. TARBP proteins
contain domains that bind double-stranded RNA [15]. TARBP1
functions as a methyltransferase that recruits an Ago protein into
RISC [16]. Additionally, TARBP2 loads the miRNA into RISC
and exhibits a double-stranded RNA (dsRNA) binding site, which
holds the miRNA inside RISC [17, 18].
In addition to loading RNAs into RISC, TRBPs also stabilize
Dicer during pre-miRNA processing. After an incorporated miRNA
binds to an mRNA, the Ago protein either cleaves the mRNA for
degradation, which is usually associated with Ago 2, or the protein
MicroRNA Expression: Protein Participants in MicroRNA Regulation 31

Fig. 3 Translational regulation through seed matching. Cartoon showing the seed region of a mature miRNA
incorporated into RISC recognizing and binding to the seed match region of an mRNA. RISC takes the mRNA to
the p-body region where it represses translation or cleaves the mRNA for degradation

in conjunction with RISC prevents translation from occurring until


the message is degraded or the message is released by RISC through
stress-induced pathways and is recycled [19] (see Fig. 3).

1.5 SiRNAs and RITS SiRNAs, or small interfering RNAs, are noncoding RNAs with
comparable size and function to miRNAs. SiRNAs differ from
miRNAs in several aspects including the pathways from transcrip-
tion to maturity, and in that while miRNAs generally regulate
mRNA networks, siRNAs typically regulate a specific target gene
[17]. RITS, or RNA-induced initiation of transcriptional silencing,
is a complex of proteins in conjunction with a mature siRNA that
inhibits the transcription of specific genes by triggering hetero-
chromatin assembly in centromeric regions.
SiRNA and RITS complexes have been experimentally charac-
terized in fission yeast. In fission yeast the RITS complex consists
of Ago1; Chp1, which is a hetero-chromatin-associated protein;
and Tas3, a novel protein that is necessary for H3-K9 methylation
[20]. This protein complex, in conjunction with a mature siRNA,
targets specific regions of DNA and silences them using methyla-
tion and heterochromatin biogenesis [21]. Upon being loaded
32 Valeria M. King and Glen M. Borchert

Fig. 4 DNA methylation facilitated by RITS and siRNA is a cartoon depicting dsRNA or double-stranded RNA
being cleaved by Dicer and matured into an siRNA or small interfering RNA. RITS is composed of at least three
protein components including Ago1, Chp1, and Tas3. The RITS protein complex, in conjunction with the siRNA,
binds to a centromeric region of DNA specified by the siRNA and enables methylation of the H3 histone by Clr4,
an H3 methyltransferase

into Ago1, a siRNA will bind a complementary centromeric region


of DNA in complex with the RITS proteins. Then K9 methylation
of the H3 histone is initiated by Clr4 protein, a histone H3 meth-
yltransferase [22]. In response to this methylation, targeted DNA
coils tightly around its associated histones thereby preventing tran-
scription of the region (see Fig. 4).

2 MiRNA Regulation

Importantly, miRNAs are themselves regulated through several


different mechanisms such as transcriptional regulations, single
nucleotide polymorphisms (SNPs), RNA editing, miRNA tailing,
and miRNA degradation [23]. MiRNA regulation can alter miRNA
expressions and protein targets, affect miRNA dosing and miRNA
proliferation, and induce miRNA degradation.

2.1 Regulation Transcriptional regulation of miRNAs is perhaps the most well-­


of MicroRNA understood mechanism by which to regulate miRNA biogenesis
Transcription [24]. MiRNA locus methylation is the untemplated addition of
MicroRNA Expression: Protein Participants in MicroRNA Regulation 33

methyl groups to a region of DNA in order to encourage tight


heterochromatin folding of DNA that prevents transcription.
MiRNAs can be down regulated when regions in the DNA that
code for the miRNA and regions that allow the miRNA to be tran-
scribed such as a promoter or transcription factor are silenced by
gene inaccessibility [25]. Hypomethylation plays a role in up-­
regulated levels of miRNAs in the cell.
Regions that are normally inaccessible to transcription can be
“opened up” by mechanisms that are as yet not fully understood.
Hepatocellular carcinoma (HCC) is associated with the dysregula-
tion of miRNAs by hypermethylating regions of the DNA which
include tumor-suppressing miRNAs and hypomethylating regions
that are normally constitutively transcribed and causes the up-­
regulation of miRNAs that can promote tumor growth [26].
Additionally, transcription can also be affected by levels of par-
ticular proteins (typically transcription factors) in the cell. For
instance, mir-145 has been shown to produce an apoptotic effect
in cells following an activation of TP53, a tumor suppressor. The
production of TP53 also stimulates the transcription of miRNA-
­145 creating an apoptosis-promoting loop. Conversely, miRNA-
­145 under-expression is observed in a variety of cancers including
breast, colon, and lung cancers [27].
Further, transcriptional regulation of miRNAs has been shown
to play a major role in embryonic stem cell (ES cell) differentiation
[28]. Four transcription factors have been implicated in regulating
miRNA levels in the cell to induce differentiation in mice: Oct4,
Sox2, Nanog, and Tcf3. Using CHIP-seq data and intensive
miRNA promoter mapping, Marson et al. were able to show that
the transcription factors bind to promoter regions that are respon-
sible for the transcription of at least 81 miRNA genes and inhibit
particular miRNAs from being transcribed [29]. This process
results in the shift of ES cells from pluripotency to specificity [30].
The transcription factors bind to the promoter regions of miRNA
genes and prevent the gene from being polymerized which in turn
results in increased expression of proteins that the targeted miRNA
regulates.

2.2 Single Nucleotide Single nucleotide polymorphisms or SNPs are areas in the genome
Polymorphisms where a single nucleotide can differ between individuals due to
typically benign mutations in the DNA sequence [31]. That said,
SNPs can drastically alter miRNA activity in the cell. A SNP in the
seed sequence of a miRNA can significantly alter its targets and
allow a certain protein to go uninhibited while another protein
becomes more stringently regulated. This can cause issues when
oncogenes, for example, are no longer being targeted to be a par-
ticular miRNA, or if tumor suppressor proteins are severely
repressed following the introduction of an SNP in a miRNA that
does not normally regulate the tumor suppressor gene [23]. SNPs
34 Valeria M. King and Glen M. Borchert

can also lead to miRNA regulation when it is not localized in the


seed region. SNPs in the passenger strand or pri-miRNA stem loop
can interfere with Drosha and DICER processing and inhibit the
production of a mature miRNA [32].

2.3 RNA Editing MiRNAs can also be regulated through RNA editing. A protein
called ADAR1 can convert adenosine molecules into inosines that
preferentially form base pairs with cytosines [33]. RNA editing may
affect pri-miRNAs, pre-miRNAs, and mature miRNA sequences,
and like SNPs, these edits can alter gene targets or decrease the
affinity for Drosha or DICER affecting miRNA processing [34].
Importantly, it is estimated that 16% of human pri-miRNAs are
edited by ADAR1 [23], suggesting that ADAR editing may well
provide a largely underappreciated layer of complexity in miRNA
biogenesis. Although miRNA editing can allow the production of
several unique miRNAs with differing targets to be produced from
a single genomic locus, the effects of miRNA editing remain largely
unexplored.

2.4 MiRNA Tailing RNA tailing is the post-transcriptional addition of nucleotides to


the 3′ end of RNA. Uridylation mainly occurs in pre- and pri-­
miRNAs. For example, during embryonic stages, members of the
let-7 family are suppressed after transcription by LIN28A and its
paralogue LIN28B. These proteins bind to the terminal loop of pri-
let7 and pre-let-7 respectively, and prevent Drosha and Dicer pro-
cessing. LIN28 proteins then employ terminal uridylyl transferases
TUT4 and TUT7 to signal pre-let-7 for decay by inducing oliguri-
dylation [35, 36]. Next, DIS3L2 exonuclease targets the oligo-U
tail and degrades the miRNA. Conversely, when LIN28 is down
regulated in cells, TUT7, TUT4, and TUT2 stimulate monouri-
dylation of pre-let-7, which increases let-7 proliferation [37].
Another type of RNA tailing is adenylation, which primarily
occurs in mature miRNAs. Adenylation can result in either miRNA
stabilization or miRNA decay [38]. As examples, miRNA-122, a
hepatic miRNA, is frequently stabilized by adenylation, whereas
poxvirus polyadenylation polymerase targets host miRNAs and
adenylates them causing their degradation. It remains unclear
what causes the difference in response following miRNA polyad-
enylation [39].

2.5 MiRNA Mature miRNA degradation has been observed in several different
Degradation systems. Though it is unclear how nucleases specify targets, numer-
ous nucleases are suspected of actively degrading miRNAs in
humans [18], C. elegans [40], and mice [41]. The first reported
instance in which miRNAs were rapidly degraded was observed in
Arabidopsis thaliana, in which mature miRNAs were cleaved and
removed by an association of 3′–5′ exonucleases called small-RNA-­
degrading nuclease [42].
MicroRNA Expression: Protein Participants in MicroRNA Regulation 35

Similarly, it has been shown that viruses destabilize specific


miRNAs by using their own RNA that contains a perfectly comple-
mentary sequence [43]. For example, T cells that are infected with
herpes virus experience a rapid decrease in miRNA-27 due to viral
noncoding RNA specifically binding and destabilizing miRNA-27
[44]. Also, mouse cytomegalovirus contains RNA that specifically
binds miRNA-27 and facilitates its degradation [45].

3 Concluding Remarks

Though miRNAs genes were almost entirely overlooked until the


turn of the millennium, miRNAs have now been shown to play an
integral part in the post-transcriptional regulation of many (if not
a majority of) protein genes. What is more, our understanding of
miRNAs and their functions as regulators continues to broaden
with significant new insights continuing to be described. For
example, miRNAs were recently shown to target not only mRNAs,
but also other noncoding RNAs such as long noncoding RNAs or
lncRNAs. Linc-MD1 is a lncRNA that is expressed during myo-
blast differentiation in muscle cells. MiRNA-133 and -135 down
regulate two transcription factors that stimulate muscle-specific
gene expression, MAML1 and MEF2C respectively. Linc-MD1
competes with these transcription factors to bind the miRNAs and
allow the transcription factors to initiate cell differentiation [46].
As another example, other small RNAs have now been shown to
behave like mature miRNAs. For example, many snoRNAs, or
small nucleolar RNAs, classically thought to simply chemically
modify other RNAs, have now been reported to undergo ­alternative
processing and behave like mature miRNAs. In a recent study, spe-
cific snoRNAs were found to be processed into miRNA-like frag-
ments and direct translational repression of target genes [47].
In conclusion, while we have learned a lot about miRNA pro-
duction and regulation in a very short time, our understanding of
these molecules is still in its infancy, and exciting new revelations
undoubtedly await.

References
1. Graves P, Zeng Y (2012) Biogenesis of mam- lism. Cell Metab 4:9–12. doi:10.1016/j.cmet.
malian microRNAs: a global view. Genomics 2006.05.009
Proteomics Bioinformatics 10:239–245. 4. Nakahara K, Carthew RW (2004) Expanding
doi:10.1016/j.gpb.2012.06.004 roles for miRNAs and siRNAs in cell regula-
2. Ha M, Kim VN (2014) Regulation of tion. Curr Opin Cell Biol 16:127–133.
microRNA biogenesis. Nat Rev Mol Cell Biol doi:10.1016/j.ceb.2004.02.006
15:509–524. doi:10.1038/nrm3838 5. Lee RC, Feinbaum RL, Ambros V (1993) The
3. Krützfeldt J, Stoffel M (2006) MicroRNAs: a C. elegans heterochronic gene lin-4 encodes
new class of regulatory genes affecting metabo- small RNAs with antisense complementarity to
36 Valeria M. King and Glen M. Borchert

lin-14. Cell 75:843–854. doi:10.1016/0092- NAs reveals rapid turnover as a common prop-
8674(93)90529-Y erty of neuronal microRNAs. Cell 141:618–631.
6. Lee RC, Ambros V (2001) An extensive class doi:10.1016/j.cell.2010.03.039
of small RNAs in Caenorhabditis elegans. 19. Zhang B, Wang Q, Pan X (2007) MicroRNAs
Science 294:862–864. doi:10.1126/science. and their regulatory roles in animals and plants.
1065329 J Cell Physiol 210:279–289. doi:10.1002/
7. miRBase. http://www.mirbase.org/index. jcp.20869
shtml. Accessed 11 Oct 2015 20. Verdel A, Jia S, Gerber S et al (2004) RNAi-­
8. Li M, Marin-Muller C, Bharadwaj U et al mediated targeting of heterochromatin by the
(2009) MicroRNAs: control and loss of con- RITS complex. Science 303:672–676.
trol in human physiology and disease. World doi:10.1126/science.1093686
J Surg 33:667–684. doi:10.1007/s00268- 21. Lam JKW, Chow MYT, Zhang Y, Leung SWS
008-9836-x (2015) siRNA versus miRNA as therapeutics
9. Yue S-B, Trujillo RD, Tang Y et al (2011) Loop for gene silencing. Mol Ther Nucleic Acids
nucleotides control primary and mature miRNA 4:e252. doi:10.1038/mtna.2015.23
function in target recognition and repression. 22. Ivanova AV, Bonaduce MJ, Ivanov SV, Klar AJ
RNA Biol 8:1115–1123. doi:10.4161/ (1998) The chromo and SET domains of the
rna.8.6.17626 Clr4 protein are essential for silencing in fission
10. Roberts JT, Cooper EA, Favreau CJ et al yeast. Nat Genet 19:192–195. doi:10.1038/566
(2013) Continuing analysis of microRNA ori- 23. Cai Y, Yu X, Hu S, Yu J (2009) A brief review
gins: formation from transposable element on the mechanisms of miRNA regulation.
insertions and noncoding RNA mutations. Genomics Proteomics Bioinformatics 7:147–
Mob Genet Elements 3:e27755. doi:10.4161/ 154. doi:10.1016/S1672-0229(08)60044-3
mge.27755 24. Lee C-T, Risom T, Strauss WM (2006)
11. Roberts JT, Cardin SE, Borchert GM (2014) MicroRNAs in mammalian development. Birth
Burgeoning evidence indicates that microR- Defects Res C Embryo Today 78:129–139.
NAs were initially formed from transposable doi:10.1002/bdrc.20072
element sequences. Mob Genet Elements 25. Xhemalce B, Robson SC, Kouzarides T (2012)
4:e29255. doi:10.4161/mge.29255 Human RNA methyltransferase BCDIN3D
12. Filshtein TJ, Mackenzie CO, Dale MD et al regulates microRNA processing. Cell 151:278–
(2012) OrbId: origin-based identification of 288. doi:10.1016/j.cell.2012.08.041
microRNA targets. Mob Genet Elements 26. He X-X, Kuang S-Z, Liao J-Z et al (2015) The
2:184–192. doi:10.4161/mge.21617 regulation of microRNA expression by DNA
13. Kobayashi H, Tomari Y (2015) RISC assem- methylation in hepatocellular carcinoma. Mol
bly: coordination between small RNAs and Biosyst 11:532–539. doi:10.1039/C4MB
Argonaute proteins. Biochim Biophys Acta. 00563E
doi:10.1016/j.bbagrm.2015.08.007 27. Schanen BC, Li X (2011) Transcriptional regu-
14. Ouellet DL, Perron MP, Gobeil L-A, Plante P, lation of mammalian miRNA genes. Genomics
Provost P (2006) MicroRNAs in gene regula- 97:1–6. doi:10.1016/j.ygeno.2010.10.005
tion: when the smallest governs it all. J Biomed 28. Boyer LA, Lee TI, Cole MF et al (2005) Core
Biotechnol 2006:20. doi:10.1155/JBB/ transcriptional regulatory circuitry in human
2006/69616 embryonic stem cells. Cell 122:947–956.
15. Parker GS, Maity TS, Bass BL (2008) dsRNA doi:10.1016/j.cell.2005.08.020
binding properties of RDE-4 and TRBP reflect 29. Marson A, Levine SS, Cole MF et al (2008)
their distinct roles in RNAi. J Mol Biol 384:967– Connecting microRNA genes to the core tran-
979. doi:10.1016/j.jmb.2008.10.002 scriptional regulatory circuitry of embryonic
16. Bahubeshi A, Tischkowitz M, Foulkes WD stem cells. Cell 134:521–533. doi:10.1016/j.
(2011) miRNA processing and human cancer: cell.2008.07.020
DICER1 cuts the mustard. Sci Transl Med 30. Kanellopoulou C (2005) Dicer-deficient
3:111ps46. doi:10.1126/scitranslmed.3002493 mouse embryonic stem cells are defective in
17. Agrawal N, Dasaradhi PVN, Mohmmed A et al differentiation and centromeric silencing.
(2003) RNA interference: biology, mecha- Genes Dev 19:489–501. doi:10.1101/gad.
nism, and applications. Microbiol Mol Biol Rev 1248505
67:657–685. doi:10.1128/MMBR.67.4.657- 31. Sun G, Yan J, Noltner K et al (2009) SNPs in
685.2003 human miRNA genes affect biogenesis and
18. Krol J, Busskamp V, Markiewicz I et al (2010) function. RNA 15:1640–1651. doi:10.1261/
Characterizing light-regulated retinal microR- rna.1560209
MicroRNA Expression: Protein Participants in MicroRNA Regulation 37

32. Lee YS, Nakahara K, Pham JW et al (2004) 40. Chatterjee S, Fasler M, Büssing I, Grosshans H
Distinct roles for Drosophila Dicer-1 and (2011) Target-mediated protection of endog-
Dicer-2 in the siRNA/miRNA silencing path- enous microRNAs in C. elegans. Dev Cell
ways. Cell 117:69–81. doi:10.1016/ 20:388–396. doi:10.1016/j.devcel.2011.
S0092-8674(04)00261-2 02.008
33. Yang W, Chendrimada TP, Wang Q et al 41. Rüegger S, Großhans H (2012) MicroRNA
(2005) Modulation of microRNA processing turnover: when, how, and why. Trends
and expression through RNA editing by ADAR Biochem Sci 37:436–446. doi:10.1016/j.
deaminases. Nat Struct Mol Biol 13:13–21. tibs.2012.07.002
doi:10.1038/nsmb1041 42. Ramachandran V, Chen X (2008) Degradation
34. Blow MJ, Grocock RJ, van Dongen S et al (2006) of microRNAs by a family of exoribonucleases
RNA editing of human microRNAs. Genome in Arabidopsis. Science 321:1490–1492.
Biol 7:R27. doi:10.1186/gb-2006-7-4-r27 doi:10.1126/science.1163728
35. Ameres SL, Zamore PD (2013) Diversifying 43. Suzuki HI, Arase M, Matsuyama H et al (2011)
microRNA sequence and function. Nat Rev Mol MCPIP1 ribonuclease antagonizes dicer and
Cell Biol 14:475–488. doi:10.1038/nrm3611 terminates MicroRNA biogenesis through pre-
36. Pasquinelli AE, Reinhart BJ, Slack F et al cursor microRNA degradation. Mol Cell
(2000) Conservation of the sequence and tem- 44:424–436. doi:10.1016/j.
poral expression of let-7 heterochronic regula- molcel.2011.09.012
tory RNA. Nature 408:86–89. doi:10.1038/ 44. Cazalla D, Yario T, Steitz JA (2010) Down-­
35040556 regulation of a host microRNA by a Herpesvirus
37. Suh M-R, Lee Y, Kim JY et al (2004) Human saimiri noncoding RNA. Science 328:1563–
embryonic stem cells express a unique set of 1566. doi:10.1126/science.1187197
microRNAs. Dev Biol 270:488–498. 45. Lee S, Song J, Kim S et al (2013) Selective deg-
doi:10.1016/j.ydbio.2004.02.019 radation of host MicroRNAs by an intergenic
38. Katoh T, Sakaguchi Y, Miyauchi K et al (2009) HCMV noncoding RNA accelerates virus pro-
Selective stabilization of mammalian microR- duction. Cell Host Microbe 13:678–690.
NAs by 3′ adenylation mediated by the cyto- doi:10.1016/j.chom.2013.05.007
plasmic poly(A) polymerase GLD-2. Genes 46. Cesana M, Cacchiarelli D, Legnini I et al
Dev 23:433–438. doi:10.1101/gad.1761509 (2011) A long noncoding RNA controls mus-
39. Backes S, Shapiro JS, Sabin LR et al (2012) cle differentiation by functioning as a compet-
Degradation of host MicroRNAs by poxvirus ing endogenous RNA. Cell 147:358–369.
poly(A) polymerase reveals terminal RNA doi:10.1016/j.cell.2011.09.028
methylation as a protective antiviral mecha- 47. Patterson D (2015) A significant percentage of
nism. Cell Host Microbe 12:200–210. small nucleolar RNAs are processed into
doi:10.1016/j.chom.2012.05.019 microRNAs. Univeraity of South Alabama
Exploring the Variety of Random
Documents with Different Content
1.C. The Project Gutenberg Literary Archive Foundation (“the
Foundation” or PGLAF), owns a compilation copyright in the
collection of Project Gutenberg™ electronic works. Nearly all the
individual works in the collection are in the public domain in the
United States. If an individual work is unprotected by copyright law
in the United States and you are located in the United States, we do
not claim a right to prevent you from copying, distributing,
performing, displaying or creating derivative works based on the
work as long as all references to Project Gutenberg are removed. Of
course, we hope that you will support the Project Gutenberg™
mission of promoting free access to electronic works by freely
sharing Project Gutenberg™ works in compliance with the terms of
this agreement for keeping the Project Gutenberg™ name associated
with the work. You can easily comply with the terms of this
agreement by keeping this work in the same format with its attached
full Project Gutenberg™ License when you share it without charge
with others.

1.D. The copyright laws of the place where you are located also
govern what you can do with this work. Copyright laws in most
countries are in a constant state of change. If you are outside the
United States, check the laws of your country in addition to the
terms of this agreement before downloading, copying, displaying,
performing, distributing or creating derivative works based on this
work or any other Project Gutenberg™ work. The Foundation makes
no representations concerning the copyright status of any work in
any country other than the United States.

1.E. Unless you have removed all references to Project Gutenberg:

1.E.1. The following sentence, with active links to, or other


immediate access to, the full Project Gutenberg™ License must
appear prominently whenever any copy of a Project Gutenberg™
work (any work on which the phrase “Project Gutenberg” appears,
or with which the phrase “Project Gutenberg” is associated) is
accessed, displayed, performed, viewed, copied or distributed:
This eBook is for the use of anyone anywhere in the United
States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it away
or re-use it under the terms of the Project Gutenberg License
included with this eBook or online at www.gutenberg.org. If you
are not located in the United States, you will have to check the
laws of the country where you are located before using this
eBook.

1.E.2. If an individual Project Gutenberg™ electronic work is derived


from texts not protected by U.S. copyright law (does not contain a
notice indicating that it is posted with permission of the copyright
holder), the work can be copied and distributed to anyone in the
United States without paying any fees or charges. If you are
redistributing or providing access to a work with the phrase “Project
Gutenberg” associated with or appearing on the work, you must
comply either with the requirements of paragraphs 1.E.1 through
1.E.7 or obtain permission for the use of the work and the Project
Gutenberg™ trademark as set forth in paragraphs 1.E.8 or 1.E.9.

1.E.3. If an individual Project Gutenberg™ electronic work is posted


with the permission of the copyright holder, your use and distribution
must comply with both paragraphs 1.E.1 through 1.E.7 and any
additional terms imposed by the copyright holder. Additional terms
will be linked to the Project Gutenberg™ License for all works posted
with the permission of the copyright holder found at the beginning
of this work.

1.E.4. Do not unlink or detach or remove the full Project


Gutenberg™ License terms from this work, or any files containing a
part of this work or any other work associated with Project
Gutenberg™.

1.E.5. Do not copy, display, perform, distribute or redistribute this


electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1
with active links or immediate access to the full terms of the Project
Gutenberg™ License.

1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or
expense to the user, provide a copy, a means of exporting a copy, or
a means of obtaining a copy upon request, of the work in its original
“Plain Vanilla ASCII” or other form. Any alternate format must
include the full Project Gutenberg™ License as specified in
paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™ works
unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or providing


access to or distributing Project Gutenberg™ electronic works
provided that:

• You pay a royalty fee of 20% of the gross profits you derive
from the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”

• You provide a full refund of any money paid by a user who


notifies you in writing (or by e-mail) within 30 days of receipt
that s/he does not agree to the terms of the full Project
Gutenberg™ License. You must require such a user to return or
destroy all copies of the works possessed in a physical medium
and discontinue all use of and all access to other copies of
Project Gutenberg™ works.

• You provide, in accordance with paragraph 1.F.3, a full refund of


any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.

• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.

1.E.9. If you wish to charge a fee or distribute a Project Gutenberg™


electronic work or group of works on different terms than are set
forth in this agreement, you must obtain permission in writing from
the Project Gutenberg Literary Archive Foundation, the manager of
the Project Gutenberg™ trademark. Contact the Foundation as set
forth in Section 3 below.

1.F.

1.F.1. Project Gutenberg volunteers and employees expend


considerable effort to identify, do copyright research on, transcribe
and proofread works not protected by U.S. copyright law in creating
the Project Gutenberg™ collection. Despite these efforts, Project
Gutenberg™ electronic works, and the medium on which they may
be stored, may contain “Defects,” such as, but not limited to,
incomplete, inaccurate or corrupt data, transcription errors, a
copyright or other intellectual property infringement, a defective or
damaged disk or other medium, a computer virus, or computer
codes that damage or cannot be read by your equipment.

1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES - Except for


the “Right of Replacement or Refund” described in paragraph 1.F.3,
the Project Gutenberg Literary Archive Foundation, the owner of the
Project Gutenberg™ trademark, and any other party distributing a
Project Gutenberg™ electronic work under this agreement, disclaim
all liability to you for damages, costs and expenses, including legal
fees. YOU AGREE THAT YOU HAVE NO REMEDIES FOR
NEGLIGENCE, STRICT LIABILITY, BREACH OF WARRANTY OR
BREACH OF CONTRACT EXCEPT THOSE PROVIDED IN PARAGRAPH
1.F.3. YOU AGREE THAT THE FOUNDATION, THE TRADEMARK
OWNER, AND ANY DISTRIBUTOR UNDER THIS AGREEMENT WILL
NOT BE LIABLE TO YOU FOR ACTUAL, DIRECT, INDIRECT,
CONSEQUENTIAL, PUNITIVE OR INCIDENTAL DAMAGES EVEN IF
YOU GIVE NOTICE OF THE POSSIBILITY OF SUCH DAMAGE.

1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If you


discover a defect in this electronic work within 90 days of receiving
it, you can receive a refund of the money (if any) you paid for it by
sending a written explanation to the person you received the work
from. If you received the work on a physical medium, you must
return the medium with your written explanation. The person or
entity that provided you with the defective work may elect to provide
a replacement copy in lieu of a refund. If you received the work
electronically, the person or entity providing it to you may choose to
give you a second opportunity to receive the work electronically in
lieu of a refund. If the second copy is also defective, you may
demand a refund in writing without further opportunities to fix the
problem.

1.F.4. Except for the limited right of replacement or refund set forth
in paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.

1.F.5. Some states do not allow disclaimers of certain implied


warranties or the exclusion or limitation of certain types of damages.
If any disclaimer or limitation set forth in this agreement violates the
law of the state applicable to this agreement, the agreement shall be
interpreted to make the maximum disclaimer or limitation permitted
by the applicable state law. The invalidity or unenforceability of any
provision of this agreement shall not void the remaining provisions.

1.F.6. INDEMNITY - You agree to indemnify and hold the Foundation,


the trademark owner, any agent or employee of the Foundation,
anyone providing copies of Project Gutenberg™ electronic works in
accordance with this agreement, and any volunteers associated with
the production, promotion and distribution of Project Gutenberg™
electronic works, harmless from all liability, costs and expenses,
including legal fees, that arise directly or indirectly from any of the
following which you do or cause to occur: (a) distribution of this or
any Project Gutenberg™ work, (b) alteration, modification, or
additions or deletions to any Project Gutenberg™ work, and (c) any
Defect you cause.

Section 2. Information about the Mission


of Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new computers.
It exists because of the efforts of hundreds of volunteers and
donations from people in all walks of life.

Volunteers and financial support to provide volunteers with the


assistance they need are critical to reaching Project Gutenberg™’s
goals and ensuring that the Project Gutenberg™ collection will
remain freely available for generations to come. In 2001, the Project
Gutenberg Literary Archive Foundation was created to provide a
secure and permanent future for Project Gutenberg™ and future
generations. To learn more about the Project Gutenberg Literary
Archive Foundation and how your efforts and donations can help,
see Sections 3 and 4 and the Foundation information page at
www.gutenberg.org.

Section 3. Information about the Project


Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-profit
501(c)(3) educational corporation organized under the laws of the
state of Mississippi and granted tax exempt status by the Internal
Revenue Service. The Foundation’s EIN or federal tax identification
number is 64-6221541. Contributions to the Project Gutenberg
Literary Archive Foundation are tax deductible to the full extent
permitted by U.S. federal laws and your state’s laws.

The Foundation’s business office is located at 809 North 1500 West,


Salt Lake City, UT 84116, (801) 596-1887. Email contact links and up
to date contact information can be found at the Foundation’s website
and official page at www.gutenberg.org/contact

Section 4. Information about Donations to


the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission of
increasing the number of public domain and licensed works that can
be freely distributed in machine-readable form accessible by the
widest array of equipment including outdated equipment. Many
small donations ($1 to $5,000) are particularly important to
maintaining tax exempt status with the IRS.

The Foundation is committed to complying with the laws regulating


charities and charitable donations in all 50 states of the United
States. Compliance requirements are not uniform and it takes a
considerable effort, much paperwork and many fees to meet and
keep up with these requirements. We do not solicit donations in
locations where we have not received written confirmation of
compliance. To SEND DONATIONS or determine the status of
compliance for any particular state visit www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states where


we have not met the solicitation requirements, we know of no
prohibition against accepting unsolicited donations from donors in
such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot make


any statements concerning tax treatment of donations received from
outside the United States. U.S. laws alone swamp our small staff.

Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.

Section 5. General Information About


Project Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could be
freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose network of
volunteer support.
Project Gutenberg™ eBooks are often created from several printed
editions, all of which are confirmed as not protected by copyright in
the U.S. unless a copyright notice is included. Thus, we do not
necessarily keep eBooks in compliance with any particular paper
edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg Literary
Archive Foundation, how to help produce our new eBooks, and how
to subscribe to our email newsletter to hear about new eBooks.

You might also like