Bioinformatics in MicroRNA Research 1st Edition Coll. download pdf
Bioinformatics in MicroRNA Research 1st Edition Coll. download pdf
Bioinformatics in MicroRNA Research 1st Edition Coll. download pdf
https://ebookultra.com
https://ebookultra.com/download/bioinformatics-in-
microrna-research-1st-edition-coll/
https://ebookultra.com/download/microrna-in-regenerative-medicine-1st-
edition-chandan-k-sen/
ebookultra.com
https://ebookultra.com/download/essays-in-bioinformatics-1st-edition-
s-jelaska/
ebookultra.com
https://ebookultra.com/download/advances-in-bioinformatics-miguel-p-
rocha/
ebookultra.com
https://ebookultra.com/download/hawaii-12th-edition-coll/
ebookultra.com
Canada 13th Edition Coll.
https://ebookultra.com/download/canada-13th-edition-coll/
ebookultra.com
https://ebookultra.com/download/japan-2017-15th-edition-coll/
ebookultra.com
https://ebookultra.com/download/dont-download-6th-edition-coll/
ebookultra.com
https://ebookultra.com/download/pattern-discovery-in-bioinformatics-
theory-algorithms-1st-edition-laxmi-parida/
ebookultra.com
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
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
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
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
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.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281
Contributors
ix
x Contributors
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.
1 Introduction
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.
2.1 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
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
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
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.
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.
3.1.2 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
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.
5 Conclusions
References
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
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
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.
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
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
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
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
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
2 MiRNA Regulation
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
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.
2.5 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
3 Concluding Remarks
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.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.
• 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 comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.
1.F.
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.
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.
Most people start at our website which has the main PG search
facility: www.gutenberg.org.