Sie sind auf Seite 1von 14

Journal of Pathology

J Pathol 2010; 220: 126139


Published online 30 October 2009 in Wiley InterScience
(www.interscience.wiley.com) DOI: 10.1002/path.2638

Invited Review

Non-coding RNAs: regulators of disease


Ryan J Taft,1 Ken C Pang,2 Timothy R Mercer,1 Marcel Dinger1 and John S Mattick1 *
1 Institute for Molecular Bioscience, University of Queensland, Brisbane QLD 4072, Australia
2 Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA

*Correspondence to: Abstract


John S Mattick, Institute for
Molecular Bioscience, University For 50 years the term gene has been synonymous with regions of the genome encoding
of Queensland, Brisbane QLD mRNAs that are translated into protein. However, recent genome-wide studies have shown
4072, Australia. that the human genome is pervasively transcribed and produces many thousands of
E-mail: j.mattick@imb.uq.edu.au regulatory non-protein-coding RNAs (ncRNAs), including microRNAs, small interfering
RNAs, PIWI-interacting RNAs and various classes of long ncRNAs. It is now clear that these
No conflicts of interest were
RNAs fulfil critical roles as transcriptional and post-transcriptional regulators and as guides
declared.
of chromatin-modifying complexes. Here we review the biology of ncRNAs, focusing on the
fundamental mechanisms by which ncRNAs facilitate normal development and physiology
and, when dysfunctional, underpin disease. We also discuss evidence that intergenic regions
associated with complex diseases express ncRNAs, as well as the potential use of ncRNAs as
diagnostic markers and therapeutic targets. Taken together, these observations emphasize
the need to move beyond the confines of protein-coding genes and highlight the fact
that continued investigation of ncRNA biogenesis and function will be necessary for a
comprehensive understanding of human disease.
Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John
Received: 14 September 2009 Wiley & Sons, Ltd.
Revised: 25 September 2009
Keywords: non-coding RNAs; small RNAs; microRNAs; RNA interference; genome-wide
Accepted: 26 September 2009
association study

Introduction to all complex genetic phenomena in the eukary-


otes, including transcriptional and post-transcriptional
Over the past decade there has been an explosion gene silencing [1322], hybrid dysgenesis [15,23], X-
of large-scale genome sequencing, which has led to chromosome dosage compensation and allelic exclu-
both great insights and unexpected conundrums. Con- sion [24,25], germ cell reprogramming [26] and para-
trary to the original expectation that more complex mutation [27,28] all of which involve epigenetic
organisms would have a greater number of genes, processes (see below).
it is now clear that humans and mice have approxi-
mately the same number of protein-coding genes as
the microscopic roundworm Caenorhabditis elegans, The expanding RNA world
most of which are orthologous, and that all multicel-
lular organisms sequenced to date have fewer protein- Small regulatory RNAs
coding genes than some simple unicellar eukaryotes
[1]. An explanation for this apparent paradox comes A fundamental and general role for regulatory RNA
from two unexpected findings: (a) that biological com- in eukaryotic biology was dramatically demonstrated
plexity generally correlates with the proportion of the in the late 1990s by the finding that double-stranded
genome that is non-protein-coding [1]; and (b) that, RNA introduced into C. elegans is cleaved by the
while only 2% of the mammalian genome encodes bidentate ribonuclease Dicer into 21 nucleotide
mRNAs, the vast majority is transcribed, largely as (nt) small RNAs that induce widespread and her-
long and short non-protein-coding RNAs (ncRNAs) itable gene silencing [2931]. Although this phe-
[210]. These findings have directly challenged the nomenon, termed RNA interference (RNAi), was orig-
traditional view of RNA as simply an intermedi- inally thought to be restricted to exogenous dsRNAs,
ary between DNA and protein, and imply that the it soon became clear that plants and animals pro-
vast majority of the genome long regarded as duce a dazzling array of endogenous small interfering
junk - encodes functional RNA species that orches- RNAs (siRNAs), microRNAs (miRNAs) and PIWI-
trate the development of complex organisms [11,12]. interacting RNAs (piRNAs) [1417,3237]. Recent
Indeed it appears that RNA signalling is central work has seen this repertoire increase even further

Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
www.pathsoc.org.uk
Non-coding RNAs: regulators of disease 127

Epigenetic Genome integrity / Transcriptional Post-transcriptional Viral defence


modifications transposon defence regulation regulation

endo-siRNAs

piRNAs miRNAs
PARs
(e.g. tiRNAs) antisense lncRNA snoRNAs
sdRNA

[repeat element] [overlapping 3 UTRs]

Figure 1. Simplified representation of regulatory ncRNAs and their functions. Generalized gene models are presented in dark
grey and light orange and overlap the double-strand DNA structure (light grey), representative of the genome. Each class of
regulatory RNA is defined by a colour. Functions are ascribed to each class of ncRNA by colour below the text at the top of the
figure. Bars and arrows indicate the direction of transcription. For more detail please see the text and references therein. PARs,
promoter-associated RNAs; lncRNAs, long non-coding RNAs; miRNAs, microRNAs; snoRNAs, small nucleolar RNAs; sdRNAs,
sno-derived RNAs; endo-siRNAs, endogenous siRNAs; piRNAs, PIWI-interacting RNAs; tiRNAs, transcription initiation RNAs

to include a host of short transcripts that sit adja- non-coding transcripts (reviewed in [15,59]), which
cent to transcription start sites [38,39], including may suggest that other Dicer-independent small RNA
promoter-associated small RNAs (PASRs) [9,40] and species are still to be discovered.
transcription initiation RNAs (tiRNAs) [41], species A detailed examination of miRNA biogenesis and
that are derived from centromeres and telomeres function is beyond the scope of this work and has
[42,43], and tiny species processed from other short recently been covered in detail in several excellent
RNAs [44] (Figure 1, Table 1). Indeed, over the last reviews [16,17,60]. However, miRNA mechanisms of
decade we have witnessed a near-exponential growth action, and the autoregulatory feedback loops that
of manuscripts devoted to regulatory RNAs (Figure 2). increasingly characterize small RNA biogenesis, are
Of the classes identified to date, miRNAs, siR- well illustrated by one of the first miRNAs discov-
NAs and piRNAs, which guide effector Argonaute ered, let-7 (Figure 3). The let-7 family of miRNAs
proteins to genomic loci or target RNAs in a sequence- is highly conserved throughout the Metazoa and func-
specific manner, have been most thoroughly inves- tions as a master temporal regulator of development
tigated. In humans there are at least 700 miRNAs, and differentiation, both in early embryos and com-
hundreds of siRNAs and millions of unique piRNA plex adult tissues such as brain, in nematode, fruitfly,
sequences [1517,45], suggesting that small RNAs zebrafish and mouse [6167]. Indeed, let-7 targets
are a substantial portion of the RNA output of cells well-established cell-cycle regulators, including Cdk6
and that they comprise a diverse, widespread and and Ras [68,69]. Like the majority of miRNAs, the let-
basal regulatory system. Indeed, several recent studies 7 precursor hairpin, or pre-miRNA, is processed from
have shown that miRNAs and piRNAs are detectable a long RNA polymerase II transcript by the nuclear
in the most primitive multicellular organisms [46] RNase Drosha, which is then exported to the cytosol
and that once acquired, they are seldom, if ever, and processed to a 22 nt mature miRNA by Dicer
lost [4749]. Although exogenous siRNAs were dis- [70].
covered a decade ago, endogenous siRNAs (endo- Each of these steps of let-7 biogenesis is tightly
siRNAs) have only recently been identified in fruit- regulated (Figure 3a). For example, while differentia-
flies and mammals [5058], where their biogene- tion factors such as Notch [71] induce transcription,
sis is dependent on Dicer processing of duplexes pluripotency factors (i.e. those that support an undif-
formed by overlapping transcripts or long perfect ferentiated cellular state), such as c-Myc, repress tran-
hairpin structures. Work from other animals, includ- scriptional activation [7274]. Likewise, the pluripo-
ing nematode and fruitfly, yeast and plants, indi- tency factor LIN28 can bind to the conserved loop
cates that that these endo-siRNAs are involved in of the primary let-7 transcript (pri-let-7) to directly
anti-viral defence, transposon silencing, chromatin inhibit the Drosha cleavage steps [7577] and can
remodelling and post-transcriptional gene regulation inhibit Dicer cleavage directly or by facilitating pre-
through Argonaute-mediated cleavage of target tran- miRNA degradation [78,79]. Completing the feed-
scripts (reviewed in [16,17]). The longest small RNA back loop, let-7 targets LIN28 [75,80,81], c-Myc
class, piRNAs, which are 2530 nt in length, are [82,83] and the c-Myc-activating gene IMP-1 [80].
also largely derived from, and involved in, trans- let-7 also forms a separate overlapping loop with the
poson defence (reviewed in [15,19]) but are largely TRIMNHL family of proteins that negatively reg-
restricted to the germline, where active transposons ulate c-Myc and enhance let-7 activity [66,8486].
could severely disrupt embryogenesis. Intriguingly, in These let-7 targets are canonical, i.e. the miRNA
a departure from the Dicer biogenesis pathway that seed sequence (nucleotides 28) binds to a target
defines siRNAs and miRNAs, piRNAs are produced mRNA 3 UTR and (generally) represses translation
by successive waves of Argonaute-cleavage of long (Figure 3b).

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
128 RJ Taft et al

Table 1. Classes of non-coding RNA in mammals


NcRNA class Characteristics References

Established ncRNA classes


Long (regulatory) non-coding The broadest class, lncRNAs, encompass all non-protein-coding RNA Reviewed in [97,98]
RNAs (lncRNAs) species >200 nt, including mRNA-like ncRNAs. Their functions include
epigenetic regulation, acting as sequence-specific tethers for protein
complexes and specifying subcellular compartments or localization
Small interfering RNAs (siRNAs) Small RNAs 2122 nt long, produced by Dicer cleavage of Reviewed in [1517,227]
complementary dsRNA duplexes. siRNAs form complexes with
Argonaute proteins and are involved in gene regulation, transposon
control and viral defence
microRNAs (miRNAs) Small RNAs 22 nt long, produced by Dicer cleavage of imperfect RNA Reviewed in [16,17,70]
hairpins encoded in long primary transcripts or short introns. They
associate with Argonaute proteins and are primarily involved in
post-transcriptional gene regulation
PIWI-interacting RNAs (piRNAs) Dicer-independent small RNAs 2630 nt long, principally restricted to Reviewed in [15,17]
the germline and somatic cells bordering the germline. They associate with
PIWI-clade Argonaute proteins and regulate transposon activity and
chromatin state
Promoter-associated RNAs A general term encompassing a suite of long and short RNAs, including Reviewed in [38,39]
(PARs) promoter-associated RNAs (PASRs) and transcription initiation RNAs
(tiRNAs) that overlap promoters and TSSs. These transcripts may regulate
gene expression
Small nucleolar RNAs Traditionally viewed as guides of rRNA methylation and Reviewed in [228]
(snoRNAs) pseudouridylation. However, there is emerging evidence that they also
have gene-regulatory roles
Other recently described classes
X-inactivation RNAs (xiRNAs) Dicer-dependent small RNAs processed from duplexes of two lncRNAs, 131
Xist and Tsix, which are responsible for X-chromosome inactivation in
placental mammals
Sno-derived RNAs (sdRNAs) Small RNAs, some of which are Dicer-dependent, which are processed 229231
from small nucleolar RNAs (snoRNAs). Some sdRNAs have been shown
to function as miRNA-like regulators of translation
microRNA-offset RNAs Small RNAs 20 nt long, derived from the regions adjacent to 232,233
(moRNAs) pre-miRNAs. Their function is unknown
tRNA-derived RNAs tRNAs can be processed into small RNA species by a conserved RNase Reviewed in [44]
(angiogenin). They are able to induce translational repression
MSY2-associated RNAs MSY-RNAs are associated with the germ cell-specific DNA/RNA binding 234
(MSY-RNAs) protein MSY2. Like piRNAs, they are largely restricted to the germline and
are 2630 nt long. Their function is unknown
Telomere small RNAs Dicer-independent 24 nt RNAs principally derived from the G-rich 43
(tel-sRNAs) strand of telomeric repeats. May have a role in telomere maintenance
Centrosome-associated RNAs A class of 3442 nt small RNAs, derived from centrosomes that show 42
(crasiRNAs) evidence of guiding local chromatin modifications

However, let-7 also targets the Dicer coding [210] and that at least 80% of this transcription is
sequence (CDS) [87] (Figure 3b), consistent with exclusively associated with long non-coding RNAs
what appears to be an emerging theme of non- (lncRNAs) [9]. Although lncRNAs have frequently
canonical miRNA targets in developmentally regulated been disregarded as artifacts of chromatin remod-
genes [8892]. Additionally, let-7 was also recently elling or transcriptional noise [94,95], there is sub-
shown to regulate HMGA2, an oncofetal gene and stantial evidence to suggest that they mirror protein-
pluripotency factor, in a cell cycle-dependent manner. coding genes. Indeed, they are frequently long (gen-
HMGA2 translation is up-regulated upon cell cycle erally >2 and some >100 kb) [96], spliced and con-
arrest but inhibited in proliferating cells [93]. Taken tain canonical polyadenylation signals [97,98]. Addi-
as an example, let-7 provides a compelling illustration tionally, lncRNA promoters are bound and regulated
of the complexity of small RNA biogenesis and func- by transcriptional factors, including Oct3/4, Nanog,
tion, and points more generally towards small RNAs CREB, Sp1, c-myc, Sox2, NF-B and p53 [99102]
having a wide range of regulatory functions facilitated and epigenetically marked with specific histone mod-
by sequence-specific interactions, any of which may ifications [102,103]. Overall, there are at least tens of
malfunction to cause disease.
thousands of lncRNAs that show signatures of selec-
tion many of which, like small RNAs, are tissue
Long non-coding RNAs and developmental stage-specific [97,104108].
Genome-wide transcriptomic studies have now shown Long ncRNAs have a variety of functions, but one
that the mammalian genome is abundantly transcribed of their primary roles appears to be as epigenetic

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 129

tumour suppressor p53 mRNA, in addition to being


translated, also functions as an RNA to inhibit the
ubiquitin ligase activity of Mdm2 [119]. Indeed, there
is increasing evidence to suggest that mRNAs contain
extensive RNA structural features [120,121], raising
the possibility that, in addition to their protein-coding
functions, mRNAs may intrinsically act as regulatory
RNAs [122].
Long ncRNAs have also been implicated in
organelle biogenesis and subcellular trafficking. For
instance, the NRON lncRNA is involved in the
cytoplasmic-to-nuclear shuttling of the NFAT tran-
scription factor [123], and the lncRNA NEAT1 /MEN
/ is required for the formation of the nuclear sub-
compartment paraspeckles in differentiated cells and
regulates the movement of primate-specific mRNAs
containing inverted Alu repeats [124127]. Another
lncRNA, Gomafu, is specifically localized in a novel
nuclear domain in a subset of neurons [128].
Although long and short regulatory RNAs are typ-
Figure 2. The rise in the number of non-coding RNA publi- ically studied and classified separately, it is impor-
cations per year. The number of PubMed indexed publications tant to note that they frequently overlap both phys-
with title, abstract or keywords matching each class of ncRNA
is plotted. Records were retrieved using an in-house Perl script ically and functionally. Indeed, the dynamic inter-
and the PubMed eFetch utility. The search terms used were: play between lncRNAs and small RNAs is evi-
piRNA, PIWI RNA, PIWI-associated RNA, PIWI-interacting dent during the process of X-chromosome inac-
RNA, siRNA, small interfering RNA, endo-siRNA, endogenous tivation (XCI), which occurs in female mammals
siRNA, miRNA, microRNA, non-coding RNA, non-coding RNA,
non-protein-coding RNA, ncRNA and lincRNA. Note that 2009
to ensure dosage compensation for X-linked genes
data were only gathered through August 2009 between the sexes. The antisense lncRNAs Xist and
Tsix are not only responsible for the chromatin
modifications that maintain XCI [24,129,130], but
regulators of protein-coding gene expression [109]. also form dsRNA duplexes in vivo that are pro-
For example, Hox genes are associated with hundreds cessed by Dicer into 2542 nt X-inactivation RNAs
of lncRNAs that define domains of differential his- (xiRNAs) [131]. Given the abundance of mam-
tone methylation and RNA polymerase accessibility malian antisense transcripts with the potential to
along the spatial and temporal axes of human devel- form dsRNA structures [132,133], such relationships
opment [110]. A lncRNA transcribed from the HOXC between lncRNAs and small RNAs may be common-
locus, termed HOTAIR, regulates the chromatin methy- place.
lation state of the HOXD locus in trans through the
polycomb repressive complex PRC2 [110]. Addition-
ally, a recent study has shown that more than 20%
of long intergenic ncRNAs associate with chromatin- Non-coding RNAs in disease
modifying complexes [111], with evidence that other
lncRNAs operate to activate gene expression through Small regulatory RNAs
Trithorax-group complexes [108]. Long ncRNAs are
also frequently associated with the phenomenon of Small RNAs have roles in virtually all develop-
genomic imprinting, which ensures that one of the two mental processes, including stem cell and germline
parental alleles of certain autosomal genes are epige- maintenance, development and differentiation, tran-
netically silenced [112116]. scriptional and post-transcriptional gene silencing and
Long ncRNAs can also directly modulate gene subcellular localization (see above, and reviewed in
transcription and protein degradation. For example, [1417,20,21]). Not surprisingly, therefore, their dis-
the lncRNA Evf2 activates the homeobox transcrip- ruption has been linked to human disease. For exam-
tion factor Dlx2 and recruits it to an ultraconserved ple, miRNAs are aberrantly expressed in: liver, pan-
genomic element, which induces transcription of Dlx5 creatic, oesophageal, stomach, colon, haematopoietic,
[117]. Mutant mice lacking Evf2 show reduced num- ovarian, breast, pituitary, prostate, thyroid, testicular
bers of GABAergic interneurons early in development and brain cancers [134138]; central nervous system
and reduced synaptic inhibition in adulthood [118], disorders (e.g. schizophrenia and Alzheimers disease)
suggesting that lncRNA-dependent processes are fun- [139]; and cardiovascular disease [140,141]. MiRNAs
damental to the central nervous system. Indeed, a are also enriched at fragile sites in the human genome
large fraction of lncRNAs is expressed in very precise and are associated with oncogenic viral integration
patterns in the brain [106]. Similarly, the archetypal sites [142,143].

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
130 RJ Taft et al

Figure 3. let-7 provides a window into miRNA biogenesis and function. (a) let-7 biogenesis and gene regulation is characterized by
a series of autoregulatory feedback loops. Lines ending in bars indicate inhibitory interactions, while those terminating in arrows
indicate activating interactions. For simplicity, all let-7 family members (of which there are 11 in vertebrates) are considered as a
group. Likewise, mammalian LIN28 homologues (LIN28 and LIN28B) and TRIMNHL family members (TRIM71 and TRIM32) are
depicted as single elements within the schematic. Mature let-7 mediates its effects through a complex composed of an Argonaute
protein (grey) and GW182 (brown), which is also depicted in simplified form in the lower panel. Consistent with its expression in
late embryogenesis, the principal targets of let-7 are cell cycle regulators, oncofetal genes, pluripotency factors and components
of the miRNA biogenesis pathway. Please see the text for more detail and references for each depicted interaction. (b) A general
schematic of mRNA transcription and miRNA targeting. Canonical miRNA targets (blue) are dependent on base pairing between
nucleotides 28, the seed sequence, and the mRNA 3 UTR. Due to the short length of the seed sequence, legitimate interactions
can be abolished and illegitimate targets created by single base changes. Non-canonical targets (orange), e.g. those in coding
sequences (CDSs) or 5 UTRs, are not reliant on the seed sequence and generally show more extensive base pairing. Canonical
and non-canonical targets are depicted for let-7a:HMGA2 [235] and let-7b:Dicer [87], respectively

Similarly, loss of specific small RNA loci is asso- study has shown that a single microdeletion involv-
ciated with PraderWilli syndrome (PWS), a disor- ing several small nucleolar RNA clusters (HBII-85
der caused by the loss of imprinting on chromo- and HBII-52) results in PWS, suggesting that loss
some 15q11-q13 and characterized by hyperphagia, of small RNAs is a causal determinant of the dis-
hypogonadism and cognitive impairment. A recent ease [144]. Consistent with this hypothesis, knockout

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 131

mice lacking the relevant snoRNAs largely recapitu- LIN28 and LIN28B, negative regulators of let-7 bio-
late the PWS phenotype [145]. Interestingly, HBII-52 genesis, correlates with repression of let-7, occurs in
forms an antisense duplex with the serotonin recep- at least 15% of human malignancies and is associ-
tor 2C (5HT2C ) mRNA and negatively regulates its ated with more advanced disease states [165]. Simi-
post-transcriptional editing [146,147], strongly impli- larly, and consistent with let-7 s role in developmental
cating it in PWS-associated and autistic neurological regulation, genetic variants of the LIN28 locus have
defects [148]. Taken together, these studies suggest recently been associated with altered timing of human
that the loss of small RNA loci plays an important pubertal growth and development [166].
role in human illness. Single nucleotide polymorphisms (SNPs) in mature
Like protein-coding genes, small RNAs can func- and precursor miRNAs have been robustly associ-
tion either as activators or inhibitors of disease. Con- ated with schizophrenia and autism [167,168], and
sistent with its role as a differentiation factor, let-7 a pathogenic SNP in the seed sequence of miR-
is a well-established tumour suppressor [61,149151] 96 is responsible for progressive hearing loss [169]
whose reduced expression is associated with poor sur- (Figure 3b). A SNP in the 3 UTR of K-Ras, a
vival in human lung cancers [152]. Likewise, mir-29b well-characterized GTPase-regulated oncogene and
expression is associated with disease-free survival in target of let-7, inhibits let-7 translational suppres-
patients with ovarian serous carcinoma [153], poten- sion and results in reduced survival in oral cancers
tially due to regulation of the de novo methyl trans- [170]. Indeed, SNPs in the 3 UTRs of mRNAs
ferases Dnmt3a and Dnmt3b [154]. Indeed, altered that abolish or create target sites may be common
expression of a broad suite of miRNAs that, dependent in miRNA-associated diseases (reviewed in [171]).
on their targets can either act as tumour suppressors As examples, SNP-induced illegitimate miRNA bind-
or oncogenes (so-called oncomiRs), has been detected ing sites are associated with muscular hypertrophy in
in virtually all cancer types examined (for reviews sheep, Tourettes syndrome and cardiovascular disease
and tables of cancer-associated miRNAs and their [171173]. More generally, allele-specific polymor-
targets, see [134,151,155,156]). Similar relationships phisms in miRNA target sites have been shown to play
are apparent in cardiovascular illnesses [140,141]. a role in the tissue-specific miRNA regulation of hun-
For example, miR-92a controls functional recovery dreds of genes, suggesting that such genetic subtleties
of ischaemic tissues in mice [157], and miR-145 and may be a widespread underlying cause of individual
miR-143, which have recently been implicated in dif- phenotypic variability [174].
ferentiation of progenitors into cardiac myocytes, are
down-regulated in injured and atherosclerotic vessels Long non-coding RNAs
[158]. MicroRNAs may even play a direct role in viral
defence. A study of human T lymphocytes has shown The data gathered to date strongly implicate lncRNAs
that miR-29a targets the HIV-1 3 UTR and directs it to in the basal regulation of protein-coding genes, includ-
P bodies, where it is suppressed by the RNA-induced ing those central to normal development and oncoge-
silencing complex (RISC) [159]. nesis, at both the transcriptional (e.g. epigenetic) and
Small RNA dysregulation occurs for multiple rea- post-transcriptional (e.g. subcellular dynamics) levels,
sons and reflects the processes involved in their and an increasing number have been functionally val-
biogenesis, regulation and targeting. MicroRNA loci idated to affect different cellular and developmental
and individual components of the miRNA biogen- pathways (see [107]). It is not surprising, then, that
esis pathway are frequently lost or amplified in a the dysregulation of lncRNAs appears to be a primary
wide range of cancers [160,161], and there is now feature of many complex human diseases, including
widespread evidence that miRNAs that act as differ- leukaemia [175], colon cancer [176], prostate cancer
entiation factors (e.g. let-7, above) are globally down- [177], breast cancer [178], hepatocellular carcinoma
regulated in cancers [134,150,151]. Indeed, ovarian [175,179], psoriasis [180], ischaemic heart disease
cancer patients who show decreased expression of [181,182], Alzheimers disease [183] and spinocere-
Dicer and Drosha, the RNases involved in miRNA bellar ataxia type 8 [184].
production (see above), are associated with poor prog- In some cases, the mechanisms by which lncRNAs
noses, suboptimal surgical cytoreduction and advanced contribute to disease have been carefully dissected. For
tumour stages [162]. Likewise, a mutation resulting in example, the dsDNA-binding protein PSF constitu-
premature termination of DICER1 results in pleuropul- tively silences the proto-oncogene GAGE6. However,
monary blastoma, a rare paediatric lung tumour [163]. at least five lncRNAs can bind to PSF, which results
Consistent with these findings, studies in mice have in deactivation of PSF-induced silencing, expression
shown that mammalian systems are highly sensitive of GAGE6 and enhanced tumorigenicity [185]. Long
to Dicer activity. Complete loss of Dicer results in the ncRNAs overlapping or antisense to protein-coding
disruption of the developmental programme and early gene promoters may also contribute to oncogenesis.
embryonic lethality [164]. A transcript antisense to the p15 tumour suppressor
Elements associated with the regulation of miRNA gene, first identified in a human leukaemia, regu-
processing can also be associated with various patholo- lates the chromatin and DNA methylation status of
gies. A recent study has shown that over-expression of the p15 locus [186]. A lncRNA antisense to p21 was

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
132 RJ Taft et al

also recently shown to behave similarly [187]. These function, PISRT1, which has also been identified as a
results, combined with the observation that antisense candidate gene in a goat model of this disease [193].
transcripts are present at thousands of protein-coding The potential role of lncRNAs in long-range
genes, have led to speculation that antisense lncR- enhancer function, and therefore dysfunction, is illus-
NAs generally control the expression of their cog- trated by the Evf2 lncRNA (see above), which
nate protein-coding genes through epigenetic modifi- may contribute to split-hand/split-foot malformation
cations [188,189]. This model has profound ramifi- 1 (SHFM1). Although the region associated with
cations for our understanding of disease, particularly this developmental disorder encompasses three genes,
cancer dysregulation of a lncRNA regulating the DLX5, DLX6 and DSS1, none are directly mutated in
expression of a tumour suppressor or oncogene, and patients [194]. Instead, exhibition of the limb pheno-
not the protein-coding sequence itself, may be one of type requires the expression of both the Dlx5 and Dlx6
the hits that leads to oncogenesis. genes to be disrupted, suggesting that SHMF1 results
from the ablation of a shared regulatory element [195].
Since it is now known that Evf2 regulates the expres-
The hidden layer of non-coding variation sion of both these genes, it warrants investigation as a
candidate SHFM1 disease locus.
These examples likely represent the tip of a very big Similarly, two lncRNAs, SOX2OT and SOX2DOT,
iceberg. The same technologies that have revealed a exhibit enriched expression in the lens of eye and over-
breadth of ncRNA expression are also driving a revo- lap a known myopia susceptibility locus (Figure 4)
lution in genome sequencing that will ultimately iden- [196,197]. These transcripts, one of which is tran-
tify variations in the human genome that underpin dis- scribed from a distal ultraconserved enhancer, also
ease susceptibility and aetiology. However, given the overlaps the SOX2 gene, itself an important regula-
focus on mutations in protein-coding exons that cause tor of ocular development. Given that developmental
most of the high-penetrance simple genetic disorders, genes are significantly enriched for a proximal asso-
the variation that occurs in non-protein-coding regions ciation with lncRNAs, it may well be that the under-
of the genome has, to date, largely been ignored or at standing of developmental disorders is likely to benefit
least not been considered [107,190]. This is changing: from an appreciation of lncRNA biology. The future
the emergence of genome-wide association studies to convergence of lncRNA identification by deep RNA
identify variant loci affecting complex diseases and sequencing with the increased resolution of disease
traits and an increased awareness of ncRNA biology variants afforded by genomic sequencing will, we sug-
have prompted a reconsideration of the underlying gest, prove a potent combination in elucidating the
protein-centric assumptions and provided a number functional contribution of ncRNAs to disease.
of novel insights into disease-causing mechanisms.
For example, many pathological mechanisms are now
known to involve aberrant regulation (and in many Non-coding RNAs as diagnostics
cases ncRNAs) rather than alterations to the protein- and therapeutics
coding sequences themselves. This is perhaps not sur-
prising, given that the primary engine of phenotypic The growing body of research showing that ncRNAs
radiation and higher complexity has been the expan- may be primary genetic regulators in complex animals
sion and divergence of the regulatory architecture that has led to the corresponding realization that this may
controls the deployment of protein components during make them ideal diagnostic markers. For example, in
differentiation and development [191], much of which some cases the expression profiles of miRNAs, in con-
may be embedded within ncRNAs [12,105]. Indeed, trast to those of protein-coding mRNAs, are able to
the same forces that drive evolutionary innovations accurately identify the origin of poorly differentiated
can result in deleterious variations. tumours and carcinomas [198,199]. Indeed, a signa-
Genome-wide association studies are beginning to ture of as few as 200 miRNAs may be sufficient for
identify novel ncRNAs as candidate disease-associated cancer classification [198], and it appears that some of
genes. For example, the lncRNA MIAT is associated the difficulties of early detection associated with colon
with myocardial infarction [181], and a novel lncRNA and other occult cancers may be overcome by pro-
induced by a chromosomal deletion that truncates the filing miRNAs obtained from patient serum, plasma,
polyadenylation site of the LUC7L gene [192] results saliva and tissues [200,201]. Likewise colon, lung and
in aberrant methylation and silencing of the neighbour- breast cancer prognosis is strongly associated with a
ing HBA2 gene, leading to the onset of -thalassemia. small suite of miRNAs (reviewed in [202]), suggesting
Indeed, many disease variants map far from protein- that assays designed to query ncRNAs may eventually
coding genes and, given the level of genome-wide become core components of the pathologists toolkit.
transcription, are therefore likely to interrupt lncRNAs. This will undoubtedly be facilitated by recent advance-
For example, a disease-causing 7.4 kb deletion asso- ments in massively parallel sequencing technologies
ciated with blepharophimosis syndrome occurs over [203,204], which allow rapid and sensitive profiling of
250 kb upstream from the nearest gene, FOXL2 [193], both long and short ncRNAs, and will almost certainly
and this mutation interrupts a lncRNA of unknown make personal genomics a reality in the next 5 years

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 133

Figure 4. Long non-coding RNAs, SOX2 distal overlapping transcript (SOX2DOT) and SOX2 overlapping transcript (SOX2OT)
map to the myopia susceptibility locus. A representation of these features, the SOX2 gene, and a highly conserved SOX2DOT
enhancer [236] are shown in the inset schematic representative of a region of the human genome (chr3: 182, 255, 415182, 945,
055; UCSC Genome Browser hg18). The relative expression of spliced ESTs corresponding to SOX2DOT or SOX2OT is depicted
in the lower half of the figure and shows that these transcripts are highly expressed in the lens of the eye. Adapted from Amaral
et al. [196]

[205]. The analysis and integration of this informa- liposome and nanoparticle delivery systems, and there
tion with other datasets (e.g. protein interaction and are currently multiple ongoing clinical trials targeting
genome-wide association studies) will pose a consid- age-related macular degeneration, respiratory syncytial
erable, but tractable, challenge well into the future. virus, acute renal failure, hepatocellular carcinoma and
The link between endogenous ncRNAs and dis- congenital pachyonychia, among others (reviewed in
ease, and the perfection of RNAi-based techniques to [212]).
silence genes in simple animals, has led to specula- There is also an increasing interest in RNA ther-
tion that RNA molecules can be employed as ther- apeutics that mimic or regulate miRNA activity in
apeutic agents. Indeed, it may be both easier and human cancers (reviewed in [213]). This could be
more productive to adjust the regulatory software facilitated by exogenous expression of a repressed
(i.e. ncRNAs) than to try and correct the hardware miRNA (using the same delivery systems as siRNA
(i.e. protein-coding genes). Hopes for RNA-based and therapeutics), by the introduction of antagomirs [214]
RNA-targeted therapies were bolstered by early suc- that are complementary and bind to miRNAs, or
cesses using siRNAs in human in vitro culture systems the use of sponges that contain multiple artificial
[206] and in targeting HIV-1 and human BCL2 with miRNA-binding sites [215]. Artificial expression of
siRNA-like molecules [207209]. Like gene therapy, specific miRNAs in vivo may be a powerful therapeu-
however, RNA therapeutics face considerable hur- tic mechanism, particularly given recent reports that
dles, including development of reliable delivery sys- over-expression of a single miRNA, miR-302, is capa-
tems, dosage regimes and techniques to ameliorate off- ble of inducing stemness [216,217].
target effects [210,211]. Nonetheless, multiple modes A series of recent studies has suggested that an
of administration have been developed, including viral, equally fruitful target may be gene promoters. Indeed,

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
134 RJ Taft et al

there is a growing body of work showing that exoge- 5. Carninci P, Sandelin A, Lenhard B, Katayama S, Shimokawa K,
nous small RNAs can activate or suppress transcription Ponjavic J, et al. Genome-wide analysis of mammalian promoter
architecture and evolution. Nat Genet 2006;38:626635.
by interfering with epigenetic marks and chromatin 6. Cheng J, Kapranov P, Drenkow J, Dike S, Brubaker S, Patel S,
formation, and thereby disrupting transcription initi- et al. Transcriptional maps of 10 human chromosomes at
ation [40,187,188,218225]. Moreover, siRNAs have 5-nucleotide resolution. Science 2005;308:11491154.
been shown to effectively modulate alternative splic- 7. Cloonan N, Forrest AR, Kolle G, Gardiner BB, Faulkner GJ,
ing [226], suggesting that, once viable, RNA therapeu- Brown MK, et al. Stem cell transcriptome profiling via massive-
scale mRNA sequencing. Nat Methods 2008;5:613619.
tics may have a wide diversity of possible uses. 8. Johnson JM, Edwards S, Shoemaker D, Schadt EE. Dark matter
in the genome: evidence of widespread transcription detected by
microarray tiling experiments. Trends Genet 2005;21:93102.
Conclusions 9. Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Will-
ingham AT, et al. RNA maps reveal new RNA classes
The absolute number of protein-coding genes encoded and a possible function for pervasive transcription. Science
2007;316:14841488.
by a genome is essentially static across all animals 10. Seila AC, Calabrese JM, Levine SS, Yeo GW, Rahl PB, Flynn
from simple nematodes to humans [1], indicating RA, et al. Divergent transcription from active promoters. Science
that additional genetic elements must be involved in 2008;322:18491851.
the development of the increasingly complex cellular, 11. Mattick JS. Non-coding RNAs: the architects of eukaryotic
physiological and neurological systems. Non-coding complexity. EMBO Rep 2001;2:986991.
12. Mattick JS. A new paradigm for developmental biology. J Exp
RNAs are likely candidates, as they are adaptively Biol 2007;210:15261547.
plastic, capable of regulating processes both broadly 13. Vaucheret H. Post-transcriptional small RNA pathways in plants:
and sequence-specifically, and are now known to be mechanisms and regulations. Genes Dev 2006;20:759771.
components of nearly all cellular and developmen- 14. Voinnet O. Origin, biogenesis, and activity of plant microRNAs.
tal systems. It is becoming clear that a comprehen- Cell 2009;136:669687.
15. Malone C, Hannon G. Small RNAs as guardians of the genome.
sive understanding of human biology must include Cell 2009;136:656668.
both small and large non-coding RNAs, and that it 16. Carthew RW, Sontheimer EJ. Origins and mechanisms of
is perhaps only through inclusion of these elements in miRNAs and siRNAs. Cell 2009;136:642655.
the biomedical research agenda, including studies to 17. Ghildiyal M, Zamore PD. Small silencing RNAs: an expanding
determine the mechanistic basis of the causative vari- universe. Nat Rev Genet 2009;10:94108.
18. Mette MF, Aufsatz W, van der Winden J, Matzke MA, Matzke
ations identified by genome-wide association studies,
AJ. Transcriptional silencing and promoter methylation triggered
that complex human diseases will be completely deci- by double-stranded RNA. EMBO J 2000;19:51945201.
phered. 19. Pal-Bhadra M, Bhadra U, Birchler JA. RNAi related mechanisms
affect both transcriptional and posttranscriptional transgene
silencing in Drosophila. Mol Cell 2002;9:315327.
Acknowledgements
20. Moazed D. Small RNAs in transcriptional gene silencing and
This study was supported by the Australian National Health genome defence. Nature 2009;457:413420.
and Medical Research Council, the Menzies Foundation, the 21. Buhler M, Moazed D. Transcription and RNAi in heterochro-
AustralianAmerican Fulbright Commission and the Royal matic gene silencing. Nat Struct Mol Biol 2007;14:10411048.
Australasian College of Physicians (KCP), the Australian 22. Matzke M, Kanno T, Huettel B, Daxinger L, Matzke AJ. Targets
of RNA-directed DNA methylation. Curr Opin Plant Biol
Research Council, the University of Queensland and the
2007;10:512519.
Queensland State Government (JSM). 23. Brennecke J, Malone C, Aravin A, Sachidanandam R, Stark A,
Hannon G. An epigenetic role for maternally inherited piRNAs
in transposon silencing. Science 2008;322:13871392.
Teaching materials 24. Kanduri C, Whitehead J, Mohammad F. The long and the
short of it: RNA-directed chromatin asymmetry in mammalian
PowerPoint slides of the figures from this review X-chromosome inactivation. FEBS Lett 2009;583:857864.
25. Yang PK, Kuroda MI. Noncoding RNAs and intranuclear posi-
are supplied as supporting information in the online tioning in monoallelic gene expression. Cell 2007;128:777786.
version of this article. 26. Sasaki H, Matsui Y. Epigenetic events in mammalian germ-
cell development: reprogramming and beyond. Nat Rev Genet
2008;9:129140.
References 27. Chandler VL. Paramutation: from maize to mice. Cell
2007;128:641645.
1. Taft RJ, Pheasant M, Mattick JS. The relationship between 28. Cuzin F, Grandjean V, Rassoulzadegan M. Inherited variation at
non-protein-coding DNA and eukaryotic complexity. Bioessays the epigenetic level: paramutation from the plant to the mouse.
2007;29:288299. Curr Opin Genet Dev 2008;18:193196.
2. Birney E, Stamatoyannopoulos JA, Dutta A, Guigo R, Gin- 29. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello
geras TR, Margulies EH, et al. Identification and analysis of CC. Potent and specific genetic interference by double-stranded
functional elements in 1% of the human genome by the ENCODE RNA in Caenorhabditis elegans. Nature 1998;391:806811.
pilot project. Nature 2007;447:799816. 30. Grishok A, Tabara H, Mello CC. Genetic requirements for
3. Core LJ, Waterfall JJ, Lis JT. Nascent RNA sequencing reveals inheritance of RNAi in C. elegans. Science 2000;287:24942497.
widespread pausing and divergent initiation at human promoters. 31. Voinnet O. Non-cell autonomous RNA silencing. FEBS Lett
Science 2008;322:18451848. 2005;579:58585871.
4. Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, 32. Hamilton AJ, Baulcombe DC. A species of small antisense
Maeda N, et al. The transcriptional landscape of the mammalian RNA in posttranscriptional gene silencing in plants. Science
genome. Science 2005;309:15591563. 1999;286:950952.

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 135

33. Zamore PD, Tuschl T, Sharp PA, Bartel DP. RNAi: double- 57. Tam OH, Aravin AA, Stein P, Girard A, Murchison EP, Che-
stranded RNA directs the ATP-dependent cleavage of mRNA at loufi S, et al. Pseudogene-derived small interfering RNAs reg-
21 to 23 nucleotide intervals. Cell 2000;101:2533. ulate gene expression in mouse oocytes. Nature 2008;453:
34. Hammond SM, Bernstein E, Beach D, Hannon GJ. An RNA- 534538.
directed nuclease mediates post-transcriptional gene silencing in 58. Watanabe T, Totoki Y, Toyoda A, Kaneda M, Kuramochi-
Drosophila cells. Nature 2000;404:293296. Miyagawa S, Obata Y, et al. Endogenous siRNAs from naturally
35. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class formed dsRNAs regulate transcripts in mouse oocytes. Nature
of tiny RNAs with probable regulatory roles in Caenorhabditis 2008;453:539543.
elegans. Science 2001;294:858862. 59. Klattenhoff C, Theurkauf W. Biogenesis and germline functions
36. Lee RC, Ambros V. An extensive class of small RNAs in of piRNAs. Development 2008;135:39.
Caenorhabditis elegans. Science 2001;294:862864. 60. Brodersen P, Voinnet O. Revisiting the principles of microRNA
37. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, target recognition and mode of action. Nat Rev Mol Cell Biol
Rougvie AE, et al. The 21-nucleotide let-7 RNA regulates 2009;10:141148.
developmental timing in Caenorhabditis elegans. Nature 61. Bussing I, Slack F, Grosshans H. let-7 microRNAs in develop-
2000;403:901906. ment, stem cells and cancer. Trends Mol Med 2008;14:400409.
38. Belostotsky D. Exosome complex and pervasive transcription in 62. Roush S, Slack FJ. The let-7 family of microRNAs. Trends Cell
eukaryotic genomes. Curr Opin Cell Biol 2009;21:352358. Biol 2008;18:505516.
39. Taft RJ, Kaplan CD, Simons C, Mattick JS. Evolution, biogen- 63. Grosshans H, Johnson T, Reinert KL, Gerstein M, Slack F.
esis and function of promoter-associated RNAs. Cell Cycle The temporal patterning microRNA let-7 regulates several
2009;8:23322338. transcription factors at the larval to adult transition in C. elegans.
40. Fejes-Toth K, Sotirova V, Sachidanandam R, Assaf G, Han- Dev Cell 2005;8:321330.
non GJ, Kapranov P, et al. Post-transcriptional processing gen- 64. Ding XC, Slack F, Grosshans H. The let-7 microRNA interfaces
erates a diversity of 5 -modified long and short RNAs. Nature extensively with the translation machinery to regulate cell
2009;457:10281032. differentiation. Cell Cycle 2008;7:30833090.
41. Taft RJ, Glazov EA, Cloonan N, Simons C, Stephen S, Faulkner 65. Chan S, Slack F. Ribosomal protein RPS-14 modulates let-
GJ, et al. Tiny RNAs associated with transcription start sites in 7 microRNA function in Caenorhabditis elegans. Dev Biol
animals. Nat Genet 2009;41:572578. 2009;138.
42. Carone D, Longo M, Ferreri G, Hall L, Harris M, Shook N, 66. Schulman BR, Esquela-Kerscher A, Slack FJ. Reciprocal expres-
et al. A new class of retroviral and satellite encoded small sion of lin-41 and the microRNAs let-7 and mir-125 during
RNAs emanates from mammalian centromeres. Chromosoma mouse embryogenesis. Dev Dyn 2005;234:10461054.
2009;118:113125. 67. Wulczyn FG, Smirnova L, Rybak A, Brandt C, Kwidzinski E,
43. Cao F, Li X, Hiew S, Brady H, Liu Y, Dou Y. Dicer indepen- Ninnemann O, et al. Post-transcriptional regulation of the
dent small RNAs associate with telomeric heterochromatin. RNA let-7 microRNA during neural cell specification. FASEB J
2009;15:12741281. 2007;21:415426.
44. Thompson DM, Parker R. Stressing out over tRNA cleavage. 68. Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kel-
Cell 2009;138:215219. nar K, Ovcharenko D, et al. The let-7 microRNA represses
45. Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miR- cell proliferation pathways in human cells. Cancer Res
Base: tools for microRNA genomics. Nucleic Acids Res 2007;67:77137722.
2008;36:D154158. 69. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R,
46. Grimson A, Srivastava M, Fahey B, Woodcroft B, Chiang HR, Cheng A, et al. RAS is regulated by the let-7 microRNA family.
King N, et al. Early origins and evolution of microRNAs and Cell 2005;120:635647.
Piwi-interacting RNAs in animals. Nature 2008;455:11931197. 70. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many
47. Peterson K, Dietrich M, Mcpeek M. MicroRNAs and metazoan roads to maturity: microRNA biogenesis pathways and their
macroevolution: insights into canalization, complexity, and the regulation. Nat Cell Biol 2009;11:228234.
Cambrian explosion. Bioessays 2009;31:736747. 71. Solomon A, Mian Y, Ortega-Cava C, Liu VW, Gurumurthy CB,
48. Heimberg AM, Sempere LF, Moy VN, Donoghue PC, Peter- Naramura M, et al. Upregulation of the let-7 microRNA
son KJ. MicroRNAs and the advent of vertebrate morphological with precocious development in lin-12/Notch hypermorphic
complexity. Proc Natl Acad Sci USA 2008;105:29462950. Caenorhabditis elegans mutants. Dev Biol 2008;316:191199.
49. Assis R, Kondrashov AS. Rapid repetitive element-mediated 72. Dangi-Garimella S, Yun J, Eves E, Newman M, Erkeland S,
expansion of piRNA clusters in mammalian evolution. Proc Natl Hammond SM, et al. Raf kinase inhibitory protein suppresses a
Acad Sci USA 2009;106:70797082. metastasis signalling cascade involving LIN28 and let-7 . EMBO
50. Ghildiyal M, Seitz H, Horwich MD, Li C, Du T, Lee S, et al. J 2009;28:347358.
Endogenous siRNAs derived from transposons and mRNAs in 73. Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM,
Drosophila somatic cells. Science 2008;320:10771081. et al. Widespread microRNA repression by Myc contributes to
51. Czech B, Malone CD, Zhou R, Stark A, Schlingeheyde C, tumorigenesis. Nat Genet 2008;40:4350.
Dus M, et al. An endogenous small interfering RNA pathway 74. Ioannidis P, Mahaira LG, Perez SA, Gritzapis AD, Sotiropoulou
in Drosophila. Nature 2008;453:798802. PA, Kavalakis GJ, et al. CRD-BP/IMP1 expression characterizes
52. Okamura K, Lai EC. Endogenous small interfering RNAs in cord blood CD34+ stem cells and affects c-myc and
animals. Nat Rev Mol Cell Biol 2008;9:673678. IGF-II expression in MCF-7 cancer cells. J Biol Chem
53. Kawamura Y, Saito K, Kin T, Ono Y, Asai K, Sunohara T, et al. 2005;280:2008620093.
Drosophila endogenous small RNAs bind to Argonaute 2 in 75. Rybak A, Fuchs H, Smirnova L, Brandt C, Pohl E, Nitsch R,
somatic cells. Nature 2008;453:793797. et al. A feedback loop comprising lin-28 and let-7 controls
54. Okamura K, Balla S, Martin R, Liu N, Lai EC. Two distinct pre-let-7 maturation during neural stem-cell commitment. Nat
mechanisms generate endogenous siRNAs from bidirectional Cell Biol 2008;10:987993.
transcription in Drosophila melanogaster. Nat Struct Mol Biol 76. Newman M, Thomson J, Hammond S. Lin-28 interaction with
2008;15:581590. the Let-7 precursor loop mediates regulated microRNA
55. Okamura K, Chung WJ, Ruby JG, Guo H, Bartel DP, Lai EC. processing. RNA 2008;14:15391549.
The Drosophila hairpin RNA pathway generates endogenous 77. Viswanathan S, Daley G, Gregory R. Selective blockade of
short interfering RNAs. Nature 2008;453:803806. microRNA processing by Lin28. Science 2008;320:97100.
56. Chung WJ, Okamura K, Martin R, Lai EC. Endogenous RNA 78. Heo I, Joo C, Cho J, Ha M, Han J, Kim VN. Lin28 mediates
interference provides a somatic defense against Drosophila the terminal uridylation of let-7 precursor microRNA. Mol Cell
transposons. Curr Biol 2008;18:795802. 2008;32:276284.

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
136 RJ Taft et al

79. Heo I, Joo C, Kim YK, Ha M, Yoon MJ, Cho J, et al. TUT4 in 102. Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D,
concert with Lin28 suppresses microRNA biogenesis through pre- et al. Chromatin signature reveals over a thousand highly
microRNA uridylation. Cell 2009;138:696708. conserved large non-coding RNAs in mammals. Nature
80. Boyerinas B, Park S, Shomron N, Hedegaard M, Vinther J, 2009;458:223227.
Andersen J, et al. Identification of let-7-regulated oncofetal 103. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Gian-
genes. Cancer Res 2008;68:25872591. noukos G, et al. Genome-wide maps of chromatin state in
81. Wu L, Belasco JG. Micro-RNA regulation of the mammalian lin- pluripotent and lineage-committed cells. Nature 2007;448:
28 gene during neuronal differentiation of embryonal carcinoma 553560.
cells. Mol Cell Biol 2005;25:91989208. 104. Amaral PP, Dinger ME, Mercer TR, Mattick JS. The eukaryotic
82. Sampson V, Rong N, Han J, Yang Q, Aris V, Soteropoulos P, genome as an RNA machine. Science 2008;319:17871789.
et al. MicroRNA let-7a down-regulates MYC and reverts 105. Amaral PP, Mattick JS. Noncoding RNA in development. Mamm
MYC-induced growth in Burkitt lymphoma cells. Cancer Res Genome 2008;19:454492.
2007;67:97629770. 106. Mercer TR, Dinger ME, Sunkin SM, Mehler MF, Mattick JS.
83. Kim HH, Kuwano Y, Srikantan S, Lee EK, Martindale JL, Specific expression of long noncoding RNAs in the mouse brain.
Gorospe M. HuR recruits let-7/RISC to repress c-Myc Proc Natl Acad Sci USA 2008;105:716721.
expression. Genes Dev 2009;23:17431748. 107. Mattick JS. The genetic signatures of noncoding RNAs. PLoS
84. Vella MC, Choi EY, Lin SY, Reinert K, Slack FJ. The C. elegans Genet 2009;5:e1000459.
microRNA let-7 binds to imperfect let-7 complementary sites 108. Dinger ME, Amaral PP, Mercer TR, Pang KC, Bruce SJ,
from the lin-41 3 UTR. Genes Dev 2004;18:132137. Gardiner BB, et al. Long noncoding RNAs in mouse embryonic
85. Lin Y, Hsieh L, Kuo M, Yu J, Kuo H, Lo W, et al. Human stem cell pluripotency and differentiation. Genome Res
TRIM71 and its nematode homologue are targets of let- 2008;18:14331445.
7 microRNA and its zebrafish orthologue is essential for 109. Mattick JS, Amaral PP, Dinger ME, Mercer TR, Mehler MF.
development. Mol Biol Evol 2007;24:25252534. RNA regulation of epigenetic processes. Bioessays 2009;31:
86. Schwamborn JC, Berezikov E, Knoblich JA. The TRIMNHL 5159.
protein TRIM32 activates microRNAs and prevents self-renewal 110. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brug-
in mouse neural progenitors. Cell 2009;136:913925. mann SA, et al. Functional demarcation of active and silent chro-
87. Forman JJ, Legesse-Miller A, Coller HA. A search for conserved matin domains in human HOX loci by noncoding RNAs. Cell
sequences in coding regions reveals that the let-7 microRNA 2007;129:13111323.
targets Dicer within its coding sequence. Proc Natl Acad Sci USA 111. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea
2008;105:1487914884. Morales D, et al. Many human large intergenic noncoding RNAs
88. Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroRNAs associate with chromatin-modifying complexes and affect gene
to Nanog, Oct4 and Sox2 coding regions modulate embryonic expression. Proc Natl Acad Sci USA 2009;106:1166711672.
stem cell differentiation. Nature 2008;455:11241128. 112. Brannan CI, Dees EC, Ingram RS, Tilghman SM. The product
89. Lal A, Kim H, Abdelmohsen K, Kuwano Y, Pullmann R, of the H19 gene may function as an RNA. Mol Cell Biol
Srikantan S, et al. p16INK4a translation suppressed by miR-24. 1990;10:2836.
PLoS ONE 2008;3:e1864. 113. Mancini-Dinardo D, Steele SJ, Levorse JM, Ingram RS, Tilgh-
90. Duursma AM, Kedde M, Schrier M, le Sage C, Agami R. miR- man SM. Elongation of the Kcnq1ot1 transcript is required
148 targets human DNMT3b protein coding region. RNA for genomic imprinting of neighboring genes. Genes Dev
2008;14:872877. 2006;20:12681282.
91. Shen WF, Hu YL, Uttarwar L, Passegue E, Largman C. 114. Redrup L, Branco MR, Perdeaux ER, Krueger C, Lewis A,
MicroRNA-126 regulates HOXA9 by binding to the homeobox. Santos F, et al. The long noncoding RNA Kcnq1ot1 organises
Mol Cell Biol 2008;28:46094619. a lineage-specific nuclear domain for epigenetic gene silencing.
92. Rigoutsos I. New tricks for animal microRNAs: targeting of Development 2009;136:525530.
amino acid coding regions at conserved and nonconserved sites. 115. Chamberlain SJ, Brannan CI. The PraderWilli syndrome
Cancer Res 2009;69:32453248. imprinting center activates the paternally expressed murine Ube3a
93. Vasudevan S, Tong Y, Steitz JA. Switching from repression antisense transcript but represses paternal Ube3a. Genomics
to activation: microRNAs can up-regulate translation. Science 2001;73:316322.
2007;318:19311934. 116. Bastepe M, Frohlich LF, Linglart A, Abu-Zahra HS, Tojo K,
94. Brosius J. Waste not, want not transcript excess in Ward LM, et al. Deletion of the NESP55 differentially
multicellular eukaryotes. Trends Genet 2005;21:287288. methylated region causes loss of maternal GNAS imprints and
95. Struhl K. Transcriptional noise and the fidelity of initiation by pseudohypoparathyroidism type Ib. Nat Genet 2005;37:2527.
RNA polymerase II. Nat Struct Mol Biol 2007;14:103105. 117. Feng J, Bi C, Clark BS, Mady R, Shah P, Kohtz JD. The Evf-2
96. Furuno M, Pang KC, Ninomiya N, Fukuda S, Frith MC, Bult C, noncoding RNA is transcribed from the Dlx-5/6 ultraconserved
et al. Clusters of internally primed transcripts reveal novel long region and functions as a Dlx-2 transcriptional coactivator. Genes
noncoding RNAs. PLoS Genet 2006;2:e37. Dev 2006;20:14701484.
97. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: 118. Bond AM, Vangompel MJ, Sametsky EA, Clark MF, Savage JC,
insights into functions. Nat Rev Genet 2009;10:155159. Disterhoft JF, et al. Balanced gene regulation by an embryonic
98. Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: brain ncRNA is critical for adult hippocampal GABA circuitry.
functional surprises from the RNA world. Genes Dev Nat Neurosci 2009;12:10201027.
2009;23:14941504. 119. Candeias MM, Malbert-Colas L, Powell DJ, Daskalogianni C,
99. Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, et al. Maslon MM, Naski N, et al. p53 mRNA controls p53 activity by
The Oct4 and Nanog transcription network regulates pluripotency managing Mdm2 functions. Nat Cell Biol 2008;10:10981105.
in mouse embryonic stem cells. Nat Genet 2006;38:431440. 120. Watts JM, Dang KK, Gorelick RJ, Leonard CW, Bess JW Jr,
100. Impey S, McCorkle SR, Cha-Molstad H, Dwyer JM, Yochum Swanstrom R, et al. Architecture and secondary structure of an
GS, Boss JM, et al. Defining the CREB regulon: a genome- entire HIV-1 RNA genome. Nature 2009;460:711716.
wide analysis of transcription factor regulatory regions. Cell 121. Warden CD, Kim SH, Yi SV. Predicted functional RNAs within
2004;119:10411054. coding regions constrain evolutionary rates of yeast proteins.
101. Cawley S, Bekiranov S, Ng HH, Kapranov P, Sekinger EA, PLoS One 2008;3:e1559.
Kampa D, et al. Unbiased mapping of transcription factor binding 122. Dinger ME, Pang KC, Mercer TR, Mattick JS. Differentiating
sites along human chromosomes 21 and 22 points to widespread protein-coding and noncoding RNA: challenges and ambiguities.
regulation of noncoding RNAs. Cell 2004;116:499509. PLoS Comput Biol 2008;4:e1000176.

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 137

123. Willingham AT, Orth AP, Batalov S, Peters EC, Wen BG, Aza- 146. Kishore S, Stamm S. The snoRNA HBII-52 regulates alternative
Blanc P, et al. A strategy for probing the function of noncoding splicing of the serotonin receptor 2C. Science 2006;311:230232.
RNAs finds a repressor of NFAT. Science 2005;309:15701573. 147. Doe CM, Relkovic D, Garfield AS, Dalley JW, Theobald DE,
124. Sunwoo H, Dinger ME, Wilusz JE, Amaral PP, Mattick JS, Humby T, et al. Loss of the imprinted snoRNA mbii-52 leads to
Spector DL. MEN / nuclear-retained non-coding RNAs are increased 5htr2c pre-RNA editing and altered 5HT2CR-mediated
up-regulated upon muscle differentiation and are essential behaviour. Hum Mol Genet 2009;18:21402148.
components of paraspeckles. Genome Res 2009;19:347359. 148. Nakatani J, Tamada K, Hatanaka F, Ise S, Ohta H, Inoue K,
125. Sasaki YT, Ideue T, Sano M, Mituyama T, Hirose T. MEN / et al. Abnormal behavior in a chromosome-engineered mouse
noncoding RNAs are essential for structural integrity of nuclear model for human 15q1113 duplication seen in autism. Cell
paraspeckles. Proc Natl Acad Sci USA 2009;106:25252530. 2009;137:12351246.
126. Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, 149. Brueckner B, Stresemann C, Kuner R, Mund C, Musch T,
Chess A, et al. An architectural role for a nuclear noncoding Meister M, et al. The human let-7a-3 locus contains an
RNA: NEAT1 RNA is essential for the structure of paraspeckles. epigenetically regulated microRNA gene with oncogenic
Mol Cell 2009;33:717726. function. Cancer Res 2007;67:14191423.
127. Chen LL, Carmichael GG. Altered nuclear retention of mRNAs 150. Peter ME. let-7 and miR-200 microRNAs: guardians against
containing inverted repeats in human embryonic stem cells: pluripotency and cancer progression. Cell Cycle 2009;8:843852.
functional role of a nuclear noncoding RNA. Mol Cell 151. Medina PP, Slack F. MicroRNAs and cancer: an overview. Cell
2009;35:467478. Cycle 2008;7:24852492.
128. Sone M, Hayashi T, Tarui H, Agata K, Takeichi M, Naka- 152. Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H,
gawa S. The mRNA-like noncoding RNA Gomafu constitutes Endoh H, et al. Reduced expression of the let-7 microRNAs in
a novel nuclear domain in a subset of neurons. J Cell Sci human lung cancers in association with shortened postoperative
2007;120:24982506. survival. Cancer Res 2004;64:37533756.
129. Wutz A, Gribnau J. X inactivation Xplained. Curr Opin Genet 153. Flavin R, Smyth P, Barrett C, Russell S, Wen H, Wei J, et al.
Dev 2007;17:387393. miR-29b expression is associated with disease-free survival in
130. Morey C, Navarro P, Debrand E, Avner P, Rougeulle C, Clerc P. patients with ovarian serous carcinoma. Int J Gynecol Cancer
The region 3 to Xist mediates X chromosome counting 2009;19:641647.
and H3 Lys-4 dimethylation within the Xist gene. EMBO J 154. Takada S, Berezikov E, Choi YL, Yamashita Y, Mano H.
2004;23:594604. Potential role of miR-29b in modulation of Dnmt3a and Dnmt3b
131. Ogawa Y, Sun BK, Lee JT. Intersection of the RNA interference expression in primordial germ cells of female mouse embryos.
and X-inactivation pathways. Science 2008;320:13361341. RNA 2009;15:15071514.
132. Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, 155. Croce CM. Causes and consequences of microRNA dysregulation
Nakamura M, et al. Antisense transcription in the mammalian in cancer. Nat Rev Genet 2009;10:704714.
transcriptome. Science 2005;309:15641566. 156. Garzon R, Calin GA, Croce CM. MicroRNAs in cancer. Annu
133. He Y, Vogelstein B, Velculescu VE, Papadopoulos N, Kin- Rev Med 2009;60:167179.
zler KW. The antisense transcriptomes of human cells. Science 157. Bonauer A, Carmona G, Iwasaki M, Mione M, Koyanagi M,
2008;322:18551857. Fischer A, et al. MicroRNA-92a controls angiogenesis and
134. Visone R, Croce CM. MiRNAs and cancer. Am J Pathol functional recovery of ischemic tissues in mice. Science
2009;174:11311138. 2009;324:17101713.
135. Pang J, Kwok W, Chen Z, Ng H. Oncogenic role of microRNAs 158. Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU,
in brain tumors. Acta Neuropathol 2009;117:599611. Muth AN, et al. miR-145 and miR-143 regulate smooth muscle
136. Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, cell fate and plasticity. Nature 2009;460:705710.
Nagel R, et al. A genetic screen implicates miRNA-372 and 159. Nathans R, Chu C, Serquina AK, Lu C, Cao H, Rana TM.
miRNA-373 as oncogenes in testicular germ cell tumors. Cell Cellular microRNA and P bodies modulate hostHIV-1
2006;124:11691181. interactions. Mol Cell 2009;34:696709.
137. Khoshnaw S, Green A, Powe D, Ellis I. MicroRNA involvement 160. Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Gian-
in the pathogenesis and management of breast cancer. J Clin nakakis A, et al. MicroRNAs exhibit high frequency genomic
Pathol 2009;62:422428. alterations in human cancer. Proc Natl Acad Sci USA
138. Novakova J, Slaby O, Vyzula R, Michalek J. MicroRNA involve- 2006;103:91369141.
ment in glioblastoma pathogenesis. Biochem Biophys Res Com- 161. Duan S, Mi S, Zhang W, Dolan ME. Comprehensive analysis of
mun 2009;386:15. the impact of SNPs and CNVs on human microRNAs and their
139. Kocerha J, Kauppinen S, Wahlestedt C. MicroRNAs in CNS regulatory genes. RNA Biol 2009; 6:epub ahead of print [PMID:
disorders. Neuromol Med 2009;111. 19458495].
140. Barringhaus K, Zamore P. MicroRNAs: regulating a change of 162. Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA,
heart. Circulation 2009;119:22172224. Schmandt R, et al. Dicer, Drosha, and outcomes in patients with
141. Sen CK, Gordillo GM, Khanna S, Roy S. Micromanaging ovarian cancer. N Engl J Med 2008;359:26412650.
vascular biology: tiny microRNAs play big band. J Vasc Res 163. Hill DA, Ivanovich J, Priest JR, Gurnett CA, Dehner LP,
2009;46:527540. Desruisseau D, et al. DICER1 mutations in familial pleuropul-
142. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, monary blastoma. Science 2009;325:965.
Yendamuri S, et al. Human microRNA genes are frequently 164. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H,
located at fragile sites and genomic regions involved in cancers. Li MZ, et al. Dicer is essential for mouse development. Nat Genet
Proc Natl Acad Sci USA 2004;101:29993004. 2003;35:215217.
143. Makunin I, Pheasant M, Simons C, Mattick JS. Orthologous 165. Viswanathan SR, Powers JT, Einhorn W, Hoshida Y, Ng TL,
microRNA genes are located in cancer-associated genomic Toffanin S, et al. Lin28 promotes transformation and is
regions in human and mouse. PLoS ONE 2007;2:e1133. associated with advanced human malignancies. Nat Genet
144. Sahoo T, Del Gaudio D, German J, Shinawi M, Peters S, 2009;41:843848.
Person R, et al. PraderWilli phenotype caused by paternal 166. Ong K, Elks C, Li S, Zhao J, Luan J, Andersen L, et al. Genetic
deficiency for the HBII-85 C/D box small nucleolar RNA cluster. variation in LIN28B is associated with the timing of puberty. Nat
Nat Genet 2008;40:719721. Genet 2009;41:729733.
145. Ding F, Li H, Zhang S, Solomon N, Camper S, Cohen P, et al. 167. Sun G, Yan J, Noltner K, Feng J, Li H, Sarkis D, et al. SNPs
SnoRNA Snord116 (Pwcr1/MBII-85) deletion causes growth in human miRNA genes affect biogenesis and function. RNA
deficiency and hyperphagia in mice. PLoS ONE 2008;3:e1709. 2009;15:16401651.

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
138 RJ Taft et al

168. Feng J, Sun G, Yan J, Noltner K, Li W, Buzin C, et al. Evidence activation and suppression in human cells. PLoS Genet
for X-chromosomal schizophrenia associated with microRNA 2008;4:e1000258.
alterations. PLoS ONE 2009;4:e6121. 188. Morris KV. Role of RNA in the regulation of gene expression.
169. Menca A, Modamio-Hybjr S, Redshaw N, Morn M, Mayo- Nutr Rev 2008;66:S31S32.
Merino F, Olavarrieta L, et al. Mutations in the seed region of 189. Morris KV. Non-coding RNAs, epigenetic memory and the
human miR-96 are responsible for nonsyndromic progressive passage of information to progeny. RNA Biol 2009;6:242247.
hearing loss. Nat Genet 2009;41:609613. 190. Visel A, Rubin EM, Pennacchio LA. Genomic views of distant-
170. Christensen BC, Moyer BJ, Avissar M, Ouellet LG, Plaza SL, acting enhancers. Nature 2009;461:199205.
McClean MD, et al. A let-7 microRNA-binding site polymor- 191. Carroll SB. Evo-devo and an expanding evolutionary synthesis: a
phism in the KRAS 3 UTR is associated with reduced survival genetic theory of morphological evolution. Cell 2008;134:2536.
in oral cancers. Carcinogenesis 2009;30:10031007. 192. Tufarelli C, Stanley JA, Garrick D, Sharpe JA, Ayyub H,
171. Chen K, Song F, Calin GA, Wei Q, Hao X, Zhang W. Polymor- Wood WG, et al. Transcription of antisense RNA leading to gene
phisms in microRNA targets: a gold mine for molecular epidemi- silencing and methylation as a novel cause of human genetic
ology. Carcinogenesis 2008;29:13061311. disease. Nat Genet 2003;34:157165.
172. Abelson JF, Kwan KY, ORoak BJ, Baek DY, Stillman AA, 193. DHaene B, Attanasio C, Beysen D, Dostie J, Lemire E, Bouchard
Morgan TM, et al. Sequence variants in SLITRK1 are associated P, et al. Disease-causing 7.4 kb cis-regulatory deletion disrupting
with Tourettes syndrome. Science 2005;310:317320. conserved non-coding sequences and their interaction with the
173. Clop A, Marcq F, Takeda H, Pirottin D, Tordoir X, Bibe B, et al. FOXL2 promotor: implications for mutation screening. PLoS
A mutation creating a potential illegitimate microRNA target site Genet 2009;5:e1000522.
in the myostatin gene affects muscularity in sheep. Nat Genet 194. Merlo GR, Paleari L, Mantero S, Genova F, Beverdam A,
2006;38:813818. Palmisano GL, et al. Mouse model of split hand/foot malforma-
174. Kim J, Bartel D. Allelic imbalance sequencing reveals that tion type I. Genesis 2002;33:97101.
single-nucleotide polymorphisms frequently alter microRNA- 195. Crackower MA, Scherer SW, Rommens JM, Hui CC, Poorkaj P,
directed repression. Nat Biotechnol 2009;27:472477. Soder S, et al. Characterization of the split hand/split foot
175. Calin GA, Liu CG, Ferracin M, Hyslop T, Spizzo R, Sevig- malformation locus SHFM1 at 7q21.3-q22.1 and analysis of a
nani C, et al. Ultraconserved regions encoding ncRNAs are candidate gene for its expression during limb development. Hum
altered in human leukemias and carcinomas. Cancer Cell Mol Genet 1996;5:571579.
2007;12:215229. 196. Amaral PP, Neyt C, Wilkins SJ, Askarian-Amiri ME, Sunkin
176. Pibouin L, Villaudy J, Ferbus D, Muleris M, Prosperi MT, SM, Perkins AC, et al. Complex architecture and regulated
expression of the Sox2ot locus during vertebrate development.
Remvikos Y, et al. Cloning of the mRNA of overexpression in
RNA 2009;15:in press.
colon carcinoma-1: a sequence overexpressed in a subset of colon
197. Andrew T, Maniatis N, Carbonaro F, Liew SH, Lau W, Spec-
carcinomas. Cancer Genet Cytogenet 2002;133:5560.
tor TD, et al. Identification and replication of three novel
177. Fu X, Ravindranath L, Tran N, Petrovics G, Srivastava S.
myopia common susceptibility gene loci on chromosome 3q26
Regulation of apoptosis by a prostate-specific and prostate
using linkage and linkage disequilibrium mapping. PLoS Genet
cancer-associated noncoding gene, PCGEM1 . DNA Cell Biol
2008;4:e1000220.
2006;25:135141.
198. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D,
178. Guffanti A, Iacono M, Pelucchi P, Kim N, Solda G, Croft LJ,
et al. MicroRNA expression profiles classify human cancers.
et al. A transcriptional sketch of a primary human breast cancer
Nature 2005;435:834838.
by 454 deep sequencing. BMC Genomics 2009;10:163.
199. Rosenfeld N, Aharonov R, Meiri E, Rosenwald S, Spector Y,
179. Lin R, Maeda S, Liu C, Karin M, Edgington TS. A large
Zepeniuk M, et al. MicroRNAs accurately identify cancer tissue
noncoding RNA is a marker for murine hepatocellular
origin. Nat Biotechnol 2008;26:462469.
carcinomas and a spectrum of human carcinomas. Oncogene 200. Cortez MA, Calin GA. MicroRNA identification in plasma and
2007;26:851858. serum: a new tool to diagnose and monitor diseases. Expert Opin
180. Sonkoly E, Bata-Csorgo Z, Pivarcsi A, Polyanka H, Kenderessy- Biol Ther 2009;9:703711.
Szabo A, Molnar G, et al. Identification and characterization of 201. Aslam MI, Taylor K, Pringle JH, Jameson JS. MicroRNAs
a novel, psoriasis susceptibility-related noncoding RNA gene, are novel biomarkers of colorectal cancer. Br J Surg
PRINS . J Biol Chem 2005;280:2415924167. 2009;96:702710.
181. Ishii N, Ozaki K, Sato H, Mizuno H, Saito S, Takahashi A, et al. 202. Swanton C, Caldas C. Molecular classification of solid tumours:
Identification of a novel non-coding RNA, MIAT, that confers risk towards pathway-driven therapeutics. Br J Cancer 2009;100:
of myocardial infarction. J Hum Genet 2006;51:10871099. 15171522.
182. Pasmant E, Laurendeau I, Heron D, Vidaud M, Vidaud D, 203. Mardis ER. Next-generation DNA sequencing methods. Annu
Bieche I. Characterization of a germ-line deletion, including Rev Genom Hum Genet 2008;9:387402.
the entire INK4/ARF locus, in a melanoma-neural system 204. Pettersson E, Lundeberg J, Ahmadian A. Generations of sequenc-
tumor family: identification of ANRIL, an antisense noncoding ing technologies. Genomics 2009;93:105111.
RNA whose expression coclusters with ARF. Cancer Res 205. Tucker T, Marra M, Friedman JM. Massively parallel sequenc-
2007;67:39633969. ing: the next big thing in genetic medicine. Am J Hum Genet
183. Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, 2009;85:142154.
Morgan TE, et al. Expression of a noncoding RNA is elevated in 206. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K,
Alzheimers disease and drives rapid feed-forward regulation of Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA inter-
-secretase. Nat Med 2008;14:723730. ference in cultured mammalian cells. Nature 2001;411:494498.
184. Daughters RS, Tuttle DL, Gao W, Ikeda Y, Moseley ML, 207. Novina CD, Murray MF, Dykxhoorn DM, Beresford PJ, Riess J,
Ebner TJ, et al. RNA gain-of-function in spinocerebellar ataxia Lee SK, et al. siRNA-directed inhibition of HIV-1 infection. Nat
type 8. PLoS Genet 2009;5:e1000600. Med 2002;8:681686.
185. Li L, Feng T, Lian Y, Zhang G, Garen A, Song X. Role of 208. Lee NS, Dohjima T, Bauer G, Li H, Li MJ, Ehsani A, et al.
human noncoding RNAs in the control of tumorigenesis. Proc Expression of small interfering RNAs targeted against HIV-1 rev
Natl Acad Sci USA 2009;106:1295612961. transcripts in human cells. Nat Biotechnol 2002;20:500505.
186. Yu W, Gius D, Onyango P, Muldoon-Jacobs K, Karp J, Fein- 209. Futami T, Miyagishi M, Seki M, Taira K. Induction of apoptosis
berg AP, et al. Epigenetic silencing of tumour suppressor gene in HeLa cells with siRNA expression vector targeted against bcl-
p15 by its antisense RNA. Nature 2008;451:202206. 2. Nucl Acids Res Suppl 2002;251252.
187. Morris K, Santoso S, Turner A, Pastori C, Hawkins P, Mcmanus 210. Grimm D, Streetz KL, Jopling CL, Storm TA, Pandey K,
M. Bidirectional transcription directs both transcriptional gene Davis CR, et al. Fatality in mice due to oversaturation

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Non-coding RNAs: regulators of disease 139

of cellular microRNA/short hairpin RNA pathways. Nature 224. Schwartz JC, Younger ST, Nguyen NB, Hardy DB, Monia BP,
2006;441:537541. Corey DR, et al. Antisense transcripts are targets for activating
211. Kleinman M, Yamada K, Takeda A, Chandrasekaran V, Nozaki small RNAs. Nat Struct Mol Biol 2008;15:842848.
M, Baffi J, et al. Sequence- and target-independent angiogenesis 225. Napoli S, Pastori C, Magistri M, Carbone GM, Catapano CV.
suppression by siRNA via TLR3. Nature 2008;452:591597. Promoter-specific transcriptional interference and c-myc gene
212. Whitehead K, Langer R, Anderson DG. Knocking down bar- silencing by siRNAs in human cells. EMBO J 2009;28:
riers: advances in siRNA delivery. Nat Rev Drug Discov 17081719.
2009;8:129138. 226. Allo M, Buggiano V, Fededa JP, Petrillo E, Schor I, de la
213. Wang V, Wu W. MicroRNA-based therapeutics for cancer. Mata M, et al. Control of alternative splicing through siRNA-
BioDrugs 2009;23:1523. mediated transcriptional gene silencing. Nat Struct Mol Biol
214. Krutzfeldt J, Rajewsky N, Braich R, Rajeev K, Tuschl T, 2009;16:717724.
Manoharan M, et al. Silencing of microRNAs in vivo with 227. Brodersen P, Voinnet O. The diversity of RNA silencing
antagomirs. Nature 2005;438:685689. pathways in plants. Trends Genet 2006;22:268280.
215. Ebert MS, Neilson J, Sharp P. MicroRNA sponges: competitive 228. Matera AG, Terns RM, Terns MP. Non-coding RNAs: lessons
inhibitors of small RNAs in mammalian cells. Nat Meth from the small nuclear and small nucleolar RNAs. Nat Rev Mol
2007;4:721726. Cell Biol 2007;8:209220.
216. Lee NS, Kim JS, Cho WJ, Lee MR, Steiner R, Gompers A, et al. 229. Taft RJ, Glazov EA, Lassmann T, Hayashizaki Y, Carninci P,
miR-302b maintains stemness of human embryonal carcinoma Mattick JS. Small RNAs derived from snoRNAs. RNA
cells by post-transcriptional regulation of Cyclin D2 expression. 2009;15:12331240.
Biochem Biophys Res Commun 2008;377:434440. 230. Saraiya AA, Wang CC. snoRNA, a novel precursor of microRNA
217. Lin SL, Chang DC, Chang-Lin S, Lin CH, Wu DT, Chen DT, in Giardia lamblia. PLoS Pathog 2008;4:e1000224.
et al. Mir-302 reprograms human skin cancer cells into a 231. Ender C, Krek A, Friedlander MR, Beitzinger M, Weinmann L,
pluripotent ES-cell-like state. RNA 2008;14:21152124. Chen W, et al. A human snoRNA with microRNA-like functions.
218. Han J, Kim D, Morris KV. Promoter-associated RNA is required Mol Cell 2008;32:519528.
for RNA-directed transcriptional gene silencing in human cells. 232. Shi W, Hendrix D, Levine M, Haley B. A distinct class of small
Proc Natl Acad Sci USA 2007;104:1242212427. RNAs arises from pre-miRNA-proximal regions in a simple
219. Hawkins PG, Santoso S, Adams C, Anest V, Morris KV. Pro- chordate. Nat Struct Mol Biol 2009;16:183189.
moter targeted small RNAs induce long-term transcriptional gene 233. Langenberger D, Bermudez-Santana C, Hertel J, Hoffmann S,
silencing in human cells. Nucleic Acids Res 2009;37:29842995. Khaitovich P, Stadler PF. Evidence for human microRNA-
220. Janowski B, Huffman K, Schwartz J, Ram R, Hardy D, Shames offset RNAs in small RNA sequencing data. Bioinformatics
D, et al. Inhibiting gene expression at transcription start sites 2009;25:22982301.
in chromosomal DNA with antigene RNAs. Nat Chem Biol 234. Xu M, Medvedev S, Yang J, Hecht NB. MIWI-independent
2005;1:216222. small RNAs (MSY-RNAs) bind to the RNA-binding protein,
221. Janowski B, Huffman K, Schwartz J, Ram R, Nordsell R, MSY2, in male germ cells. Proc Natl Acad Sci USA
Shames D, et al. Involvement of AGO1 and AGO2 in 2009;106:1237112376.
mammalian transcriptional silencing. Nat Struct Mol Biol 235. Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ,
2006;13:787792. et al. Combinatorial microRNA target predictions. Nat Genet
222. Janowski B, Younger S, Hardy D, Ram R, Huffman K, Corey D. 2005;37:495500.
Activating gene expression in mammalian cells with promoter- 236. Pennacchio LA, Ahituv N, Moses AM, Prabhakar S,
targeted duplex RNAs. Nat Chem Biol 2007;3:166173. Nobrega MA, Shoukry M, et al. In vivo enhancer analysis
223. Morris K. Small interfering RNA-induced transcriptional gene of human conserved non-coding sequences. Nature 2006;444:
silencing in human cells. Science 2004;305:12891292. 499502.

J Pathol 2010; 220: 126139 DOI: 10.1002/path


Copyright 2009 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Das könnte Ihnen auch gefallen