JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY
VOL. 67, NO. 10, 2016
ª 2016 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION
ISSN 0735-1097/$36.00
PUBLISHED BY ELSEVIER
http://dx.doi.org/10.1016/j.jacc.2015.12.051
THE PRESENT AND FUTURE STATE-OF-THE-ART REVIEW
Long Noncoding RNAs From Clinical Genetics to Therapeutic Targets? Reinier A. Boon, PHD,a,b Nicolas Jaé, PHD,a Lesca Holdt, MD, PHD,b,c Stefanie Dimmeler, PHDa,b
ABSTRACT Recent studies suggest that the majority of the human genome is transcribed, but only about 2% accounts for proteincoding exons. Long noncoding RNAs (lncRNAs) constitute a heterogenic class of RNAs that includes, for example, intergenic lncRNAs, antisense transcripts, and enhancer RNAs. Moreover, alternative splicing can lead to the formation of circular RNAs. In support of putative functions, GWAS for cardiovascular diseases have shown predictive singlenucleotide polymorphisms in lncRNAs, such as the 9p21 susceptibility locus that encodes the lncRNA antisense noncoding RNA in the INK4 locus (ANRIL). Many lncRNAs are regulated during disease. For example, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and myocardial infarction-associated transcript (MIAT) were shown to affect endothelial cell functions and diabetic retinopathy, whereas lincRNA-p21 controls neointima formation. In the heart, several lncRNAs were shown to act as microRNA sponges and to control ischemia-reperfusion injury or act as epigenetic regulators. In this review, the authors summarize the current understanding of lncRNA functions and their role as biomarkers in cardiovascular diseases. (J Am Coll Cardiol 2016;67:1214–26) © 2016 by the American College of Cardiology Foundation.
I
Listen to this manuscript’s
n the early days of molecular biology, the catego-
was solved stepwise by the discovery of intronic
rization of ribonucleic acids (RNAs) was rela-
sequences (4,5) and various functional noncoding
tively
RNAs, such as the RNA component of RNaseP (6),
straightforward,
dividing
RNAs
into
protein-coding messenger RNAs (mRNAs) and a
small nuclear RNAs (7), longer noncoding transcripts
reasonable number of functional noncoding RNAs,
(e.g., Xist and H19 [8,9]), and microRNAs (miRNAs)
such as transfer RNAs or ribosomal RNAs. However,
(10) (Figure 1A). Meanwhile, the biogenesis and func-
by comparing the sizes of haploid genomes (C values)
tion of many of these noncoding RNAs is well
from different organisms, it was soon discovered that
described. MiRNAs, for example, are small cyto-
these values do not necessarily correlate with devel-
plasmic RNAs of 20 to 23 nt in length exhibiting an
opmental complexity (1). The assumption was that
important gene-regulatory potential. They are tran-
the majority of the genome is noncoding DNA or
scribed as primary transcripts, processed to precur-
“junk” DNA (2). Nevertheless, this supposed non-
sors in the nucleus, and subsequently exported to
functionality was already questioned at this time (1),
the cytoplasm to be diced into mature double-
and subsequent RNA-driven hybridization reactions
stranded miRNAs. Of this duplex, 1 strand is incorpo-
with nonrepetitive sea urchin DNA revealed that so-
rated into RNA-induced silencing complexes to
called heterogeneous nuclear RNA covers a larger
silence gene expression at the post-transcriptional
portion of the genome than mRNA isolated from pol-
level (see He et al. [11] for a detailed review). In
ysomal fractions (3). This obvious imbalance between
contrast, small nuclear RNAs are part of small nuclear
RNA being transcribed and known RNA species
ribonucleoproteins, which participate in the dynamic
audio summary by JACC Editor-in-Chief Dr. Valentin Fuster. From the aInstitute for Cardiovascular Regeneration, University of Frankfurt, Frankfurt, Germany; bGerman Center of Cardiovascular Research, Berlin, Germany; and the cInstitute of Laboratory Medicine, Ludwig-Maximilians-University Munich, Munich, Germany. Drs. Boon, Jaé, and Dimmeler have filed for a patent regarding long noncoding RNAs in cardiovascular disease. Dr. Holdt has reported that she has no relationships relevant to the contents of this paper to disclose. Manuscript received October 13, 2015; revised manuscript received November 26, 2015, accepted December 14, 2015.
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
1215
Long Noncoding RNAs in Cardiovascular Disease
assembly of the spliceosome on nascent pre-mRNAs
methyl-CpG-binding domain protein 1 to re-
ABBREVIATIONS
and the process of splicing (12). Finally, with the
cruit H3K9 methyltransferases to its own
AND ACRONYMS
advent of next-generation sequencing technologies,
imprinted gene network, thus establishing
it became evident that the human genome is nearly
repressive chromatin marks, which, in turn,
pervasively transcribed, which means that the major-
control gene expression of the given network
ity of its bases are associated with at least 1 primary
members (29). An additional function of
transcript (13,14); however, protein-coding exons ac-
lncRNAs is their involvement in develop-
count for only about 2% of the genome (15,16).
mental differentiation. The lncRNA Neat1, for
Today,
long
noncoding
RNAs
(lncRNAs)
are
example, is expressed in a broad range of
ANRIL = antisense noncoding RNA in the INK4 locus
APF = autophagy-promoting factor
Bvht = Braveheart CAD = coronary artery disease CARL = cardiac-apoptosis
frequently defined as non-protein-coding transcripts
tissues
larger than 200 nt to distinguish them from small
cell
noncoding RNAs (17); however, their classification is
Neat1 is implicated in the assembly of para-
not standardized. Accordingly, lncRNA nomenclature
speckles (nuclear subdomains of unclear
CDKN2B = cyclin-dependent
relies on diverse empirical features, for example,
function) by recruiting paraspeckle proteins,
kinase inhibitor 2B
origin of transcription, tissue specificity, molecular
as well as in the maintenance of these dy-
CHRF = cardiac hypertrophy
function, or mechanism of action (see Ma et al. [18]
namic nuclear structures (32).
and
is
differentiation
up-regulated (30,31).
upon
Functionally,
for a detailed review). Regarding their genomic
The detailed mechanism of action is
localization, lncRNAs can be transcribed from inter-
closely linked with lncRNA function. Those
related long noncoding RNA
Cas9 = CRISPR-associated protein 9
related factor
circRNA = circular RNA CRISPR = clustered regularly interspaced short palindromic
genic regions (long intervening noncoding RNAs),
lncRNAs implicated in the regulation of
from within introns of protein-coding genes (intronic
chromatin states often recruit chromatin-
lncRNAs), or from the antisense strand of a given
modifying complexes to genes in the vicin-
association studies
lncRNA = long noncoding RNA
repeats
GWAS = genome-wide
gene (natural antisense transcripts) (19). Interest-
ity of their own transcription (so-called cis
ingly, the primary sequence of lncRNAs is only poorly
regulation). A prominent example is the
conserved; however, this circumstance is partially
lncRNA antisense noncoding RNA in the INK4
associated lung
compensated by a higher degree of structural con-
locus (ANRIL), which is transcribed close to
adenocarcinoma transcript 1
servation
the genomic locus of INK4A and interacts
Frequently,
(20)
to
lncRNAs
maintain
from
with components of the polycomb repressive complex 1 (PRC1) to induce silencing of
genes in different species), indicating a putative
INK4A (33). Beyond being restricted to their
functional relation, despite the previously mentioned
sites of transcription, lncRNAs can also re-
sequence divergence (22). Back-splicing of exons,
cruit epigenetic complexes to distant chro-
thereby forming circular RNAs (circRNAs), can also
mosomal sites in order to regulate gene
generate lncRNAs (23,24). In contrast to the previ-
expression (so-called trans regulation). For
ously
better
example, the lncRNA HOTAIR, which is
conserved and are more stable because of their
transcribed from the HOXC locus, represses
resistance to exonucleases (24,25).
transcription in trans across 40 kb of the
lncRNAs,
also
transcribed
(21).
conserved genomic regions (same order of flanking
described
are
functionality
circRNAs
are
LncRNA molecular functions are very diverse
HOXD locus (34). Besides their well-known
MALAT1 = metastasis-
MDRL = mitochondrial dynamic related long noncoding RNA
Mhrt = myosin heavy chainassociated RNA transcript
MIAT = myocardial RNA transcript
miRNA = micro–RNA mRNA = messenger RNA NFIA = nuclear factor IA PRC1 = polycomb repressive complex 1
PRC2 = polycomb repressive complex 2
(Central Illustration). One of the best-studied aspects
influence on transcription and chromatin
is their role in the epigenetic regulation of allelic
states, lncRNAs can also act as molecular
endothelial cell-enriched
expression. A generic model for this is the well-
scaffolds to organize protein complexes or,
migration/differentiation-
known process of X chromosome dosage compensa-
conversely, as decoys or sinks for specific
tion in female mammals. X chromosome inactivation
target molecules to limit their availability.
is achieved by expression of the lncRNA Xist, which
For instance, HOTAIR is, in addition to its
coats 1 X chromosome and acts as a scaffold for the
epigenetic function, known to act at the post-
recruitment of silencing factors (e.g., polycomb
translational level by serving as assembly
repressive complex 2 [PRC2]) (26,27). Related to X
platform for protein ubiquitination (35), whereas the
chromosome inactivation is the developmental pro-
lncRNA PANDA inhibits the expression of apoptotic
cess of genomic imprinting, which results in the
genes by sequestering the transcription factor NF-YA,
exclusive expression of specific genes from only 1
thereby acting as molecular sink (36). Another
parent chromosome (28). In this context, the highly
example of an lncRNA acting as a molecular decoy is
expressed long intervening noncoding RNA H19,
lncRNA-ATB, which competitively binds miR-200 to
now known to be part of an imprinted gene network,
sequester this miRNA from its mRNA targets ZEB1/2,
was among the first imprinted genes identified (9).
thereby promoting prometastatic functions of trans-
Recent studies suggest that H19 interacts with
forming growth factor– b (37). Finally, lncRNAs are
SENCR = smooth muscle and
associated long noncoding RNA
shRNA = small hairpin RNA siRNA = small interfering RNA SNP = single-nucleotide polymorphism
1216
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
F I G U R E 1 Noncoding RNAs of the Human Transcriptome
Human genome
mRNAs
Translation
Other noncoding RNAs*
Noncoding transcriptome
Long noncoding RNAs
miRNAs piRNAs snoRNAs snRNAs tRNAs H1 RNA RMRP 7SL RNA TERC scaRNAs rRNAs Y RNAs
Long noncoding RNAs (lncRNAs) Long intervening noncoding RNAs (lincRNAs) Intronic long noncoding RNAs (lncRNAs) Natural antisense transcripts (NATs) Enhancer RNAs (eRNAs) Circular RNAs (circRNAs)
Post-transcriptional gene regulation Germ line transposon silencing RNA modification pre-mRNA splicing Translation pre-tRNA processing mtRNA processing, rRNA processing Direction of protein traffic Maintenance of telomere ends RNA modification Translation DNA replication, small RNA maturation
The human genome is nearly pervasively transcribed, resulting in an almost noncoding transcriptome with various RNA species. *Depicted RNA classification does not rely on a strict 200 nt cutoff. H1 RNA ¼ RNA component of ribonuclease P; miRNAs ¼ microRNAs; mRNA ¼ messenger RNA; mtRNA ¼ mitochondrial RNA; piRNAs ¼ piwi-interacting RNAs; RMRP ¼ RNA component of RNase MRP; rRNA ¼ ribosomal RNA; scaRNAs ¼ small Cajal body-specific RNAs; 7SL RNA ¼ signal recognition particle RNA; snoRNAs ¼ small nucleolar RNAs; snRNAs ¼ small nuclear RNAs; TERC ¼ telomerase RNA component; tRNAs ¼ transfer RNAs; Y RNAs ¼ part of the RoRNP.
involved in several steps of post-transcriptional
IDENTIFICATION OF NONCODING RNAs AS HOT SPOTS
gene regulation, including mRNA processing and
FOR SINGLE-NUCLEOTIDE POLYMORPHISMS: LESSONS
mRNA stability, as well as translation. In this context,
TO
mRNA processing can be altered through 2 different
STUDIES. Genome-wide association studies (GWAS)
mechanisms. First, natural antisense transcripts can
have
form RNA-RNA duplexes with their corresponding
morphisms (SNPs) in noncoding regions may have
sense transcripts, thereby altering splice-site recog-
functional consequences. SNPs in 3 0 untranslated
nition and spliceosome recruitment, a mechanism
regions of mRNAs may interfere with miRNA binding
observed, for example, during processing of the first
or other regulatory processes that control mRNA
intron of neuroblastoma MYC mRNA (38). Second,
stability or translation (44). For example, it has
lncRNAs can indirectly regulate splicing, as seen for
been
metastasis-associated lung
tran-
disease–associated variation in the 3 0 untranslated
script 1 (MALAT1), which regulates splicing factor
region of the basic-helix-loop-helix transcription
phosphorylation and can interact with U1 small nu-
factor TCF21, which critically controls coronary artery
clear RNA, a component of the spliceosome (39,40).
smooth muscle cell and cardiac fibroblast maturation,
adenocarcinoma
LEARN
FROM
suggested
recently
GENOME-WIDE
that
reported
ASSOCIATION
single-nucleotide
that
a
coronary
poly-
heart
With respect to mRNA stability, lncRNAs are impli-
disrupts a miR-224 binding site (45). Variations in
cated in processes leading to stabilization, as well as
miRNAs can themselves alter mRNA targeting and
destabilization, of a given transcript. An interesting
may thereby dysregulate the gene expression pat-
example is the lncRNA BACE1AS, which forms an
terns controlled by the miRNA. SNPs may also affect
RNA-RNA duplex with BACE1 mRNA, thus preventing
the expression of lncRNAs. In addition, variations
miR-485-5p-mediated
(41).
may affect the structure of lncRNAs by interfering
Reciprocally, the long intervening noncoding RNA H19
with RNA folding and the formation of RNA structures
was shown to coordinate the interaction of protein K
or by modulating protein-RNA interactions. Although
homology-type splicing regulatory proteins and labile
little is known regarding SNPs in noncoding regions,
repression
of
BACE1
transcripts in the cytoplasm, which favors protein K
the first hints that SNPs in noncoding regions may
homology-type splicing regulatory protein–mediated
indeed be relevant were provided in 2007 (46–48).
destabilization (42). Finally, lncRNAs can also exert
Among the 58 genomic loci (p < 107) of athero-
their effects at the level of protein synthesis,
sclerosis susceptibility identified by GWAS (49),
as seen for the mouse lncRNA Uchl1AS, which is
the most significant genetic locus was found at
required for the association of Uchl mRNA with active
chromosome 9p21. SNPs in this region have been
polysomes (43).
associated with different endpoints of atherosclerotic
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
CENTRAL I LLU ST RAT ION
Long Noncoding RNAs in Cardiovascular Disease
The RNA World and Cardiovascular Disease: Long Noncoding RNAs
Boon, R.A. et al. J Am Coll Cardiol. 2016; 67(10):1214–26.
Depending on the subcellular localization, long noncoding RNAs (lncRNAs) can act via various mechanisms. By mimicking transcription factor binding sites, doublestranded regions of lncRNAs can bind to transcription factors, thereby functioning as a decoy. Via binding to chromatin modifiers, lncRNAs contribute to epigenetic silencing or activation of gene expression. LncRNAs that bind to exon/intron junctions of pre–messenger RNA (mRNA) can influence (alternative) splicing. Several lncRNAs act as a scaffold in ribonucleoprotein (RNP) complexes. In the cytoplasm, numerous lncRNAs have been shown to bind microRNAs (miRNAs), thereby preventing the miRNAs from binding to and regulating their mRNA targets. By masking binding sites for proteins or miRNAs, cytoplasmic lncRNAs can alter the stability of mRNAs.
cardiovascular disease (46–48,50). Chromosome 9p21
exon of ANRIL, and mechanistic studies have shown
spans a region of approximately 50 kb but does not
that the 9p21 risk allele disrupts an inhibitory STAT1
comprise protein-coding genes (51) and has therefore
binding site, leading to up-regulation of ANRIL
been considered a “gene desert.” The closest protein-
expression (52,55). ANRIL is expressed in cells and
coding genes are cyclin-dependent kinase inhibitor
tissues that are relevant in atherogenesis, and mul-
2A CDKN2A and cyclin-dependent kinase inhibitor 2B
tiple isoforms have been identified that can be clas-
(CDKN2B), which are located >100 kb from associated
sified according to their exon structures in 4 major
SNPs. Chromosome 9p21 encodes different transcripts
linear and circular spliced forms (56,57). The molec-
of the lncRNA ANRIL (also known as CDKN2B anti-
ular function of linear ANRIL has been investigated
sense RNA 1). A potential function of this lncRNA in
using RNA interference and overexpression studies
vascular disease is supported by several studies
(56,58). Targeting exon 1 (all linear ANRIL isoforms)
showing its expression to be associated with risk for
or exon 19 (long ANRIL isoforms only) by small
coronary atherosclerosis (52), carotid arteriosclerosis
interfering RNAs (siRNAs) in smooth muscle cells
(53), peripheral artery disease (54), and other vascular
reduced cell viability (58). Results from this study are
disease (see Holdt and Teupser [51] for a review). The
in line with overexpression studies in which ANRIL
top associated SNPs are found adjacent to the last
increased cellular proliferation (56). In addition,
1217
1218
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
ANRIL increased adhesion and decreased apoptosis,
through multiple let-7-binding sites (68,69), thereby
which are proatherosclerotic cellular mechanisms
preventing
(56). At the molecular level, ANRIL binds to epige-
target genes associated with various cardiovascular
netic effector proteins of PRC1 and PRC2 (33,56,59)
diseases (70).
let-7
from
inhibiting
expression
of
and recruits them to promoter regions, thereby
In summary, there is increasing evidence that
epigenetically regulating gene expression patterns
lncRNAs may control atherosclerosis risk at non-
(56,60,61). The recruitment is mediated though re-
coding regions identified by GWAS of cardiovascular
petitive Alu elements in ANRIL, which are mirrored in
disease. Apart from ANRIL at 9p21, no other lncRNAs
the promoters of its target genes (56).
at GWAS loci identified in European populations
Two other studies have investigated lncRNAs at
have thus far been functionally characterized. Results
genomic loci associated with myocardial infarction
from the first functional studies of lncRNAs from
and coronary artery disease (CAD) in Japanese and
Asian association studies hold great promise for
Chinese cohorts (62,63). Because these loci were not
novel mechanistic insights in the pathology of
replicated in GWAS performed in populations of Eu-
atherosclerosis.
ropean descent, it remains to be determined whether these loci and lncRNAs also contribute to athero-
REGULATION OF lncRNAs IN
sclerosis risk in non-Asian populations. The lncRNA
CARDIOVASCULAR DISEASE
myocardial infarction-associated transcript (MIAT) is transcribed from chromosome 22q12.1 and was iden-
Because lncRNAs can be linked to cardiovascular
tified as a risk allele for myocardial infarction in a
disease, it is highly likely that lncRNAs are also
large-scale case-control association study in Japanese
regulated in certain cardiovascular disease states.
subjects (62). The risk allele of a SNP in exon 5 was
The first evidence that lncRNAs are dynamically
shown to increase MIAT expression, likely through
regulated in the heart came from studies of cardiac
binding
development during embryogenesis in mice (71,72).
of
a
hitherto
uncharacterized
protein.
Although the function of MIAT in the heart is still
In recent years, several lncRNAs have been
unknown, MIAT expression was reduced in periph-
described
eral blood mononuclear cells isolated from patients
required for cardiac development. The first of these
as
highly
induced
during
and
even
with ST-segment elevation myocardial infarction.
lncRNAs was termed Braveheart (Bvht) (71). Bvht was
Recent studies suggest that MIAT controls endothe-
identified by screening gene expression databases
lial cell functions and also plays a role in diabetic
for lncRNAs highly expressed in the heart. Bvht does
retinopathy (discussed in later text).
not code for any protein, but Bvht depletion dimin-
In a Chinese population, variations of lncRNA H19
ished the capacity of embryonic stem cells to
at chromosome 11p15.5 have been associated with the
differentiate into cardiomyocytes. Furthermore, Bvht
risk for CAD (63). Here it was shown that subjects
was found to induce a transcriptional program
carrying 3 or 4 risk alleles at the H19 locus had a
downstream of the transcription factor MESP1, a key
significantly increased risk for CAD and higher Gen-
cardiac differentiation transcription factor. At the
sini scores than those with 0 to 2 risk alleles (63). The
same time that Bvht was described, another lncRNA,
rs2067051 polymorphism site is located in the 5 0 flank
Fendrr, was found to be required for heart develop-
of H19 and is near the imprinting control regions,
ment in mice (72). The investigators found that ge-
which are critical for regulation of H19 expression.
netic deletion of Fendrr caused ventricular defects
The second SNP in the H19 locus is located in the last
during development. Like Bvht, Fendrr regulates a
exon of H19, but how this contributes to H19 expres-
transcriptional network required for cardiac devel-
sion levels or function is unclear at the moment. H19
opment via interaction with histone modifying
is an imprinted lncRNA, which is induced during
complexes (PRC2 and TrxG/MLL). In contrast to
embryogenesis
birth,
Bvht, which does not seem to have a human homo-
except in adult skeletal muscle and heart. H19
log, Fendrr is also present in the human genome, but
expression is increased in various tumors (64).
its function during human development or cardio-
Moreover, H19 is highly expressed in the neointima of
vascular disease is not known.
and
down-regulated
after
rats 7 to 14 days after vascular injury (65) and is
Recently, 3 novel lncRNAs were identified as
reexpressed in human atherosclerotic plaques (66).
induced during differentiation of human embryonic
Genetic studies showed that polymorphisms in H19
stem cells to endothelial cells (73). These lncRNAs,
are correlated with high blood pressure (67), a known
termed TERMINATOR, ALIEN, and PUNISHER, not
risk factor for CAD. Mechanistic studies have shown
only increase in expression during differentiation to
that H19 can bind and sequester the miRNA let-7
endothelial cells, they also contribute to endothelial
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
commitment and identity. Knockdown of any of the 3
valve interstitial cells. Furthermore, HOTAIR was also
lncRNAs results in cardiovascular defects in devel-
reduced in bicuspid aortic valves in comparison with
oping zebrafish embryos.
tricuspid aortic valves, suggesting a role in valve
Studies that assessed enhancer regions that func-
calcification.
tion as transcriptional activators in the human heart
lncRNAs AS BIOMARKERS. A potential application
found that many of these regions localize to non-
for lncRNAs lies in the use of circulating lncRNA
coding parts of the genome (74,75), raising the sug-
levels as biomarkers for cardiovascular disease.
gestion that enhancer RNAs could arise from these
Several pioneering studies have shown this to be
loci that regulate cardiac gene expression. Several
feasible. In 2014, the group of Thum (84) described
other lncRNAs are also regulated during development
the mitochondrial lncRNA uc022bqs.1, which the in-
or in various cardiovascular disease models (76,77).
vestigators termed LIPCAR, as increased in the
Finally, recent studies report that circRNAs are
plasma of patients with cardiac remodeling after
expressed in endothelial cells and are regulated by
acute myocardial infarction and in patients with
hypoxia in vitro (78). Among the many circRNAs
chronic heart failure. Furthermore, plasma LIPCAR
identified, the circular form of ZNF292 (cZNF292)
levels can be used as a prognostic indicator for car-
was shown to regulate angiogenesis, and its inhibi-
diovascular mortality, although it is not clear whether
tion reduces sprouting angiogenesis and endothelial
circulating LIPCAR arises from the mitochondria or
cell proliferation (78). Although the mechanism un-
nuclei of cardiac myocytes or even from other cell
derlying the biological effects of cZNF292 are not
types (85). Another study from 2014 (86) described
fully clear, a function as a miR sponge has been
the association of whole blood cell expression
excluded (78).
levels of the lncRNAs aHIF, ANRIL, KCNQ1OT1, MIAT,
REGULATION OF lncRNAs DURING PATHOLOGY. The
and MALAT1 with acute myocardial infarction. The
first evidence that lncRNAs are differentially regu-
investigators
lated in human cardiac pathologies came from RNA
KCNQ1OT1 could be used as prognostic markers for
deep sequencing experiments by Yang et al. (79). The
left ventricular function. The studies mentioned thus
investigators were able to detect 18,480 lncRNAs in
far focused on markers for cardiac dysfunction, but
human left ventricles. This study showed that the
because (coronary) atherosclerosis is the underlying
lncRNA expression pattern distinguished between
cause of an acute ischemic insult, biomarkers that
ischemic and nonischemic failing hearts. The func-
reflect coronary atherosclerosis are also highly clini-
tion of the specifically regulated lncRNAs was not
cally relevant. Yang et al. (87) found that the lncRNA
assessed. By studying expression of lncRNAs that are
they termed Coromarker is increased in plasma from 2
regulated after myocardial infarction in mice, the
cohorts of patients with CAD compared with healthy
group of Pedrazzini (80) identified several human
control subjects. Whether Coromarker is released
homologs that are differentially expressed in heart
directly by cells in atherosclerotic plaques or rather
samples of patients with dilated cardiomyopathy
reflects the proinflammatory state of circulating cells
and aortic stenosis. The investigators hypothesized
remains to be seen. Finally, circulating levels of the
that
lncRNA Tapsaki predicted mortality of patients with
1
particular
lncRNA
(Novlnc6),
which
is
down-regulated in patients with dilated cardiomy-
further
showed
that
ANRIL
and
acute kidney injury (88).
opathy, controls cardiomyocyte identity. A descrip-
Very recent studies additionally suggest that
tive study of lncRNA expression in right heart failure
circRNAs, which are regulated in endothelial cells
identified several lncRNAs that are differentially
in vitro (78), might be potential biomarkers. Memczak
expressed in patient heart samples compared with
et al. (89) reported that circRNAs can be detected at
unused donor hearts (81). LncRNAs are likely also
higher levels in peripheral blood compared with the
regulated during progression of cardiomyopathy, and
linear host genes and might be suitable as biomarkers
expression differences among pathologies, such as
for disease. However, their regulation in cardiovas-
hypertrophic and dilated cardiomyopathy, undoubt-
cular pathologies is unclear.
edly also exist. However, to date, no studies
As with any circulating biomarker for cardiovas-
describing lncRNA expression in these human dis-
cular disease, the cellular origin of the circulating
eases have been published. An interesting study by
lncRNAs is often unclear, and it is not known
Carrion et al. (82) described the repression of a
whether these lncRNAs are causally involved in the
lncRNA, HOTAIR, previously described as controlling
pathophysiology of the underlying disease. Further-
expression of the HOXD locus (34) and enhancing
more, quantification of RNA relies, at least with
metastasis (83), by cyclic stretch in human aortic
the majority of techniques, on polymerase chain
1219
1220
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
se-
occurring during diabetes, are regulated by lncRNAs
quences, which is inherently troublesome with blood
reaction–mediated
amplification
of
specific
(Figure 2A). Particularly diabetic retinopathy, which
samples (90). Detection of linear lncRNAs, which
is characterized by pathological proliferation of
might be even more susceptible to degradation (if
retinal vessels and increased vasopermeability, can
not adequately protected) and are often expressed at
be ameliorated by interfering with lncRNA ex-
very low levels, may be more challenging compared
pression in vivo. Inhibition of the hypoxia- and
with miRNAs. Moreover, to perform an unbiased
diabetes-induced lncRNA MALAT1 (93–95) prevented
lncRNA transcriptome analysis with small volumes
diabetes-induced vascular complications (95). Specif-
of plasma or whole blood from large patient cohorts
ically,
by RNA deep sequencing could prove technically
improved retinal function and reduced retinal cell
challenging and is likely very costly. However,
death (95). MALAT1 inhibition reduced endothelial cell
because there are potentially more noncoding genes
hyperproliferation and vascular inflammation without
intraocular
injection
of
MALAT1
shRNA
than coding genes, analyzing signatures or sets
directly affecting endothelial barrier function (95,96).
of circulating lncRNAs could prove very powerful
These findings are consistent with the previously
as biomarkers for specific (cardiovascular) disease
described inhibition of endothelial cell proliferation by
states.
pharmacological gapmer-mediated MALAT1 silencing or genetic deletion of MALAT1 (93). Gapmers are
FIRST INSIGHTS INTO THE PATHOPHYSIOLOGICAL ROLE OF NONCODING RNAs IN
single-stranded oligonucleotides that consist of a DNA stretch flanked by locked nucleic acid nucleotides, inducing ribonuclease H-dependent cleavage of the
CARDIOVASCULAR DISEASE
target RNA, and showed an efficient inhibition of nuclear localized MALAT1 in vitro (93). Intraperitoneal
Whereas transcriptomic analysis showed that many
injection of gapmers furthermore efficiently sup-
lncRNAs are regulated in cardiovascular disease,
pressed MALAT1 in various organs, particularly in the
relatively little is known regarding the causal func-
intima in vivo (93). Whereas inhibition of pathological
tional contribution of lncRNAs in disease models
angiogenesis by MALAT1 silencing using shRNAs or
in vivo. To understand their biological functions,
gapmers may be useful under conditions of diabetic
lncRNAs can be genetically deleted by conventional
retinopathy, endothelial proliferation is required for
gene-targeting strategies, for example, by intro-
ischemia-induced neovascularization and inhibition
ducing a transcriptional stop signal to block RNA
of MALAT1 in this context may be detrimental (93).
polymerase (reviewed by Bassett et al. [91]). Short
Diabetic retinopathy is also controlled by the
RNAs, such as siRNAs, modified antisense oligonu-
lncRNA MIAT (97,98). MIAT knockdown by shRNA
cleotides, or gapmers can also achieve inhibition.
in rats reduced endothelial cell apoptosis and
Whereas
oligonucleotides
vascular leakage and counteracted diabetes-induced
preferentially target cytoplasmic RNAs, gapmers
up-regulation of proinflammatory proteins, thereby
introduce ribonuclease H-dependent cleavage of nu-
alleviating retinal vessel impairment (98). The study
clear RNAs. Alternatively, viral vectors can deliver
additionally suggests that MIAT is acting as a miR-150
small hairpin RNAs (shRNAs) targeting the respective
sponge to control endothelial cell functions and dia-
lncRNA. Overexpression of lncRNAs may be more
betic retinopathy.
siRNAs
and
antisense
challenging because some lncRNAs affect gene
A very recent study described the identification of
expression in their local genomic environments; as
2 lncRNAs that are induced by hypoxia in endothelial
such, conventional overexpression with plasmids or
cells and contribute to angiogenesis (99). In vitro
viral vectors may not result in appropriate localiza-
silencing of either of these 2 lncRNAs, called
tion of these lncRNAs. Alternatively, a recently
linc00323-003 and MIR503HG, results in reduction of
described strategy using clustered regularly inter-
proliferation of endothelial cells and subsequent in-
spaced short palindromic repeats (CRISPR)/CRISPR-
hibition of angiogenic sprouting. Moreover, embed-
associated protein 9 (Cas9) technology, which allows
ding endothelial cells in which linc00323-003 and
the activation of a specific promoter by using a fusion
MIR503HG are silenced in human induced pluripo-
protein of catalytically inactive Cas9 (dCas9) with the
tent
transcriptional activator domain VP64, may be used
ex vivo results in a diminished contribution of endo-
(92) to activate expression of the endogenous
thelial cells to these engineered heart tissues. Inter-
lncRNA.
estingly, both lncRNAs inhibit the expression of the
VASCULAR DISEASE. D i a b e t i c
stem
cell–derived
engineered
heart
tissue
retinopathy and
transcription factor GATA2, thereby potentially con-
a n g i o g e n e s i s . Vascular complications, such as those
trolling endothelial cell proliferation (99). Because
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
F I G U R E 2 Long Noncoding RNAs Regulate Vascular Function
Normal
Diabetic retinopathy
MIAT
VEGF
° Vascular leakage ° Cell death (in vivo)
Other mechanisms
° Pro-angiogenic signaling °
miR-150 Cell cycle genes (e.g. cyclins) MALAT 1 Normal
° Proliferation ° Cell death
p38-P
Hypoxia
° Endothelial cell migration tube formation
Other mechanisms
linc00323-003
° Pericyte loss ° Proliferation ° Migration
GATA2
° Endothelial tube formation
MIR503HG
Lipid homeostasis HDL
LDL
NFIA miR-382-5p RP5-833A20.1
ANRIL
SENCR
linc-p21 MDM2
PRC1/2 Cell viability Proliferation
MDK MYOCD PTN ACTA2 Contractile phenotype
P53 Proliferation Apoptosis
(A) The long noncoding RNAs (lncRNAs) myocardial infarction-associated transcript (MIAT) and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) have been implicated in angiogenesis and inflammation in diabetic retinopathy. Both lncRNAs are up-regulated in diabetic retinopathy and affect endothelial cell function. MALAT1 is also induced by hypoxia. Linc00323-003 and MIR503HG are both induced by hypoxia and regulate endothelial proliferation and migration via GATA2. (B) The lncRNA RP5-833A20.1 was reported to control cholesterol homeostasis, with potential implications for atherosclerosis. Antisense noncoding RNA in the INK4 locus (ANRIL), smooth muscle and endothelial cell-enriched migration/differentiation-associated long noncoding RNA (SENCR), and linc-p21 were shown to be involved in phenotypic control of cells in the vascular wall. HDL ¼ high-density lipoprotein; LDL ¼ low-density lipoprotein; NFIA ¼ nuclear factor IA; PRC1/2 ¼ PRC1, polycomb repressive complex 1/2.
1221
1222
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
of the lack of evolutionary conservation of these
cells and endothelial cells. When the investigators
lncRNAs, their contribution to angiogenesis in vivo in
inhibited SENCR in smooth muscle cells, the cells
small animal models could not be assessed; hence, the
exhibited a more promigratory and less contractile
in vivo function of these lncRNAs is unclear.
phenotype, suggesting a potential role in vascular
Neointima formation and atherosclerosis. Neointima
pathologies characterized by smooth muscle dedif-
formation is influenced by linc-p21, which regulates
ferentiation and migration, such as restenosis and
p53-dependent gene expression and thereby re-
atherosclerosis.
presses smooth muscle cell proliferation and stimu-
LncRNAs may also regulate risk factors for CAD,
lates apoptosis of vascular smooth muscle cells (100)
such as metabolism or hypertension. For example,
(Figure 2B). In vivo silencing of linc-p21 by lentiviral
liver-enriched lncLSTR was shown to regulate sys-
vector-delivered siRNAs reduced neointima forma-
temic lipid metabolism in mice (103), which may
tion after endothelial injury (100).
secondarily affect atherosclerosis.
In vitro studies suggested that the lncRNA RP5-
CARDIAC DISEASE. Several studies addressed the
833A20.1 influences cholesterol metabolism and
role of lncRNAs in cardiac hypertrophy and remodel-
atherosclerosis. RP5-833A20.1 was shown to decrease
ing (Figure 3). Wang et al. (104) identified AK017121,
nuclear factor IA (NFIA) by inducing hsa-miR-382-5p
which they named cardiac-apoptosis related lncRNA
expression, thereby reducing high-density lipopro-
(CARL), as anoxia-regulated lncRNA, which sup-
tein cholesterol but increasing low-density lipopro-
pressed apoptosis and mitochondrial fission in vitro
tein and proinflammatory cytokines. Overexpression
and reduced ischemia-reperfusion injury in vivo.
of the putative lncRNA target NFIA induced an athe-
Mechanistically, the investigators showed that CARL
roprotective lipid status and reduced atherosclerotic
binds to miR-539 and acts as a sponge to block miR-539
lesion formation in ApoE/ mice, however, the
functions in mitochondrial fission. CARL thereby
causal involvement of the lncRNA itself has not been
derepressed the miR-539 target PHB2 and prevented
demonstrated (101).
anoxia-induced mitochondrial fission. Mitochondrial
Using RNA sequencing of human coronary artery
fission was also regulated by the lncRNA mitochon-
smooth muscle cells, Bell et al. (102) identified a
drial dynamic related lncRNA (MDRL). MDRL inhibits
lncRNA, smooth muscle and endothelial cell-enriched
mitochondrial fission and apoptosis in vitro by
migration/differentiation-associated long noncoding
down-regulating miR-361, which controls processing
RNA (SENCR), enriched in vascular smooth muscle
of another miRNA, miR-484. In vivo intracoronary
F I G U R E 3 Long Noncoding RNAs Regulate Cardiac Function
Hypertrophy
Cardiomyocyte cell death miR-539 Mitochondrial fission
miR484
miR361
CARL
Mhrt
MDRL
CHRF
Brg1 Myd88
miR-489 Autophagy
ATG7
miR188-3p
APF
Novlnc6 Nkx2.5 Bmp10
Several long noncoding RNAs (lncRNAs) were shown to regulate cardiac physiology. Cardiac-apoptosis related long noncoding ribonucleic acid (CARL), mitochondrial dynamic related long noncoding ribonucleic acid (MDRL), and autophagy-promoting factor (APF) regulate cardiomyocyte cell death by inhibiting microRNAs. Cardiac hypertrophy related factor (CHRF) also functions as a microRNA sponge in the context of cardiac hypertrophy and inhibits miR-489. The lncRNA myosin heavy chain-associated ribonucleic acid transcript (Mhrt) regulates chromatin remodeling and cardiac hypertrophy. Novlnc6 is involved in cardiac development.
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
delivery of adenoviral-expressed MDRL reduced
viral gene delivery of siRNAs or overexpression of
myocardial
ischemia-reperfusion
transcripts, antisense oligonucleotides might be
injury by inhibition of mitochondrial fission and
used to block lncRNA functions. Although the ef-
apoptosis (104).
ficiency of antisense oligonucleotide delivery is
infarction
after
AK048451, which was subsequently named cardiac hypertrophy
related
factor
(CHRF),
was
lower in the heart compared with the liver, recent
highly
studies suggest that antisense miRNA inhibitors,
induced by angiotensin II in vitro and in murine
so-called antimiRs or antagomirs, can be used to
transverse aortic constriction models, as well as in
improve cardiovascular diseases in small animals
human heart failure in vivo. CHRF is widely expressed
(63,64). Some of these findings have also been
in cardiovascular cells and other tissues but has a
confirmed in more clinically relevant large animal
specific function in cardiomyocytes (105). CHRF
models (108). Anti-miRNAs have been successfully
induced cardiomyocyte hypertrophy in vitro and
and safely used in clinical phase 2 studies,
apoptosis in vivo. The mechanism of action was
although for the treatment of liver disease (109),
attributed to binding of miR-489, which derepresses
which is easier to target by therapeutic RNAs
the miR-489 target Myd88 to regulate cardiomyocyte
compared with the heart. Whether lncRNAs can be
hypertrophy
lncRNA
targeted by a similar strategy remains to be deter-
autophagy-promoting factor (APF) was reported to
mined. Gapmers might be useful, particularly for
regulate autophagic cell death in vitro, and inhibition
nuclear-expressed lncRNAs. Locked nucleic acid
of APF by siRNA reduced ischemia-reperfusion injury
gapmers have, for example, been used for targeting
in vivo in mice. APF thereby targeted miR-188-3p,
liver PCSK9 in nonhuman primates, but a phase 1
which represses the autophagy-related protein 7 (106).
clinical study was halted (110). Gapmers against
A cluster of antisense transcripts from the myosin
survivin and hypoxia-inducing factor 1a have been
heavy chain 7 locus (named myosin heavy chain-
applied weekly over as long as 1 year without
associated RNA transcripts [Mhrt]) were shown to be
safety concerns (111,112). Future approaches to
highly expressed in the murine heart (107). Mhrt is
therapeutically interfere with lncRNA functions
cardiac specific and prevents stress-induced cardiac
may also include small molecules specifically
(105).
More
recently,
the
remodeling by directly interfering with the Brg1
designed to interfere with RNA-protein complexes.
helicase, thereby inhibiting a stress-induced gene
LncRNAs exhibit diverse functions, and under-
expression program. The investigators also identified
standing the molecular mechanism mediated by a
a human ortholog of Mhrt, which is also induced and
given lncRNA may be complicated. Particularly if
could perhaps be used therapeutically to prevent
the lncRNA interferes with chromatin structure or
pathological cardiac remodeling in humans (107).
epigenetic control mechanisms, deciphering the
PROMISE AND CHALLENGES IN USING lncRNAs AS THERAPEUTIC TARGETS OR BIOMARKERS Increasing evidence suggests that lncRNAs may play crucial roles in the regulation of pathophysiological processes in the cardiovascular system and might be therapeutically be targeted. However, the field of lncRNAs is still in its infancy and faces many challenges:
exact mechanism by which the lncRNA functions is not trivial. Furthermore, some lncRNAs may act through
more
than
1
mechanism
of
action.
Although several studies carefully document that lncRNAs act as sponges for miRNAs to control cardiac functions, it is unclear whether such a mechanism can be generally considered. Because most lncRNAs are expressed at low levels, it is hard to reconcile a sponge function for highly expressed miRNAs with only 1 miRNA binding site per lncRNA. The Stoffel group recently reported that
Many lncRNAs are not conserved at the sequence
changes in the abundance of miRNA target sites are
level and are expressed only in primates. Unless
unlikely to cause significant effects (113). Although
structural conservation is established in the future,
this study only used the 3 0 untranslated region of
it will be challenging to understand the biological
mRNA, the stoichiometry of target RNA and miRNA
properties of these regulatory RNAs. Humanized
has to be carefully considered. In addition, it is
models or organoid cultures will be necessary to
thus far unclear whether bound miRNA may
screen for biological properties of primate-specific lncRNAs before embarking on costly and ethically disputed nonhuman primate studies. Therapeutically targeting lncRNAs may potentially be used to combat cardiovascular disease. Besides
instead affect lncRNA expression or functions. Mechanistic studies are complicated by the fact that lncRNAs are often expressed in many transcript variants, which may or may not have common
functions.
Like
proteins, RNAs
can
be
1223
1224
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
modified and such modifications (e.g., methyl-
Forschungsgemeinschaft (SFB834 and SFB902), the
ation, editing) may affect lncRNA functions.
European Research Council (Drs. Boon and Dimmeler),
Finally, some of the transcripts annotated as non-
and the Leducq Network “Mirvad” for support.
coding RNAs may code for proteins. For example, lncRNAs may encode short peptides that have been overlooked in the past, as recently reported for a cardiac lncRNA (114).
REPRINT REQUESTS AND CORRESPONDENCE: Prof.
Stefanie
Dimmeler,
Institute
of
Cardiovascular
Regeneration, Centre for Molecular Medicine, J. W.
ACKNOWLEDGMENTS The authors thank Rie Manavski
Goethe University Frankfurt am Main, Theodor-
for help with the graphic design and the German
Stern-Kai 7, D-60590 Frankfurt, Germany. E-mail:
Center for Cardiovascular Research, the Deutsche
[email protected].
REFERENCES 1. Thomas CA Jr. The genetic organization of chromosomes. Annu Rev Genet 1971;5:237–56.
Consortium. Initial sequencing and analysis of the human genome. Nature 2001;409:860–921.
2. Ohno S. So much “junk” DNA in our genome. Brookhaven Symp Biol 1972;23:366–70.
16. Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science 2001;
3. Hough BR, Smith MJ, Britten RJ, et al. Sequence complexity of heterogeneous nuclear RNA in sea urchin embryos. Cell 1975;5:291–9. 4. Chow LT, Gelinas RE, Broker TR, et al. An amazing sequence arrangement at the 50 ends of adenovirus 2 messenger RNA. Cell 1977;12:1–8. 5. Berget SM, Moore C, Sharp PA. Spliced segments at the 50 terminus of adenovirus 2 late mRNA. Proc Natl Acad Sci U S A 1977;74:3171–5. 6. Stark BC, Kole R, Bowman EJ, et al. Ribonuclease P: an enzyme with an essential RNA component. Proc Natl Acad Sci U S A 1978;75: 3717–21. 7. Lerner MR, Steitz JA. Antibodies to small nuclear RNAs complexed with proteins are produced by patients with systemic lupus erythematosus. Proc Natl Acad Sci U S A 1979;76:5495–9. 8. Brown CJ, Ballabio A, Rupert JL, et al. A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome. Nature 1991;349:38–44. 9. Bartolomei MS, Zemel S, Tilghman SM. Parental imprinting of the mouse H19 gene. Nature 1991; 351:153–5. 10. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993;75:843–54. 11. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 2004; 5:522–31. 12. Patel SB, Bellini M. The assembly of a spliceosomal small nuclear ribonucleoprotein particle. Nucleic Acids Res 2008;36:6482–93. 13. ENCODE Project Consortium. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007;447:799–816. 14. Djebali S, Davis CA, Merkel A, et al. Landscape of transcription in human cells. Nature 2012;489: 101–8. 15. Lander ES, Linton LM, Birren B, et al., for the International Human Genome Sequencing
29. Monnier P, Martinet C, Pontis J, et al. H19 lncRNA controls gene expression of the imprinted gene network by recruiting MBD1. Proc Natl Acad Sci U S A 2013;110:20693–8.
291:1304–51.
30. Chen LL, Carmichael GG. Altered nuclear
17. Mercer TR, Dinger ME, Mattick JS. Long noncoding RNAs: insights into functions. Nat Rev Genet 2009;10:155–9.
retention of mRNAs containing inverted repeats in human embryonic stem cells: functional role of a nuclear noncoding RNA. Mol Cell 2009;35: 467–78.
18. Ma L, Bajic VB, Zhang Z. On the classification of long non-coding RNAs. RNA Biol 2013;10: 925–33. 19. Moran VA, Perera RJ, Khalil AM. Emerging functional and mechanistic paradigms of mammalian long non-coding RNAs. Nucleic Acids Res 2012;40:6391–400. 20. Johnsson P, Lipovich L, Grandér D, et al. Evolutionary conservation of long non-coding RNAs; sequence, structure, function. Biochim Biophys Acta 2014;1840:1063–71. 21. Pang KC, Frith MC, Mattick JS. Rapid evolution of noncoding RNAs: lack of conservation does not mean lack of function. Trends Genet 2006;22:1–5. 22. Ulitsky I, Shkumatava A, Jan CH, et al. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell 2011;147:1537–50. 23. Jeck WR, Sorrentino JA, Wang K, et al. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 2012;19:141–57. 24. Memczak S, Jens M, Elefsinioti A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013;495:333–8.
31. Sunwoo H, Dinger ME, Wilusz JE, et ε/b nuclear-retained non-coding RNAs regulated upon muscle differentiation essential components of paraspeckles.
al. MEN are upand are Genome
Res 2009;19:347–59. 32. Mao YS, Sunwoo H, Zhang B, et al. Direct visualization of the co-transcriptional assembly of a nuclear body by noncoding RNAs. Nat Cell Biol 2011;13:95–101. 33. Yap KL, Li S, Muñoz-Cabello AM, et al. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell 2010;38:662–74. 34. Rinn JL, Kertesz M, Wang JK, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007;129:1311–23. 35. Yoon JH, Abdelmohsen K, Kim J, et al. Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination. Nat Commun 2013;4:2939. 36. Hung T, Wang Y, Lin MF, et al. Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet 2011;43: 621–9.
25. Cocquerelle C, Mascrez B, Hétuin D, et al. Missplicing yields circular RNA molecules. FASEB J 1993;7:155–60.
37. Li W, Kang Y. A new Lnc in metastasis: long
26. Brown CJ, Hendrich BD, Rupert JL, et al. The human XIST gene: analysis of a 17 kb inactive X-
38. Krystal GW, Armstrong BC, Battey JF. N-myc mRNA forms an RNA-RNA duplex with endogenous antisense transcripts. Mol Cell Biol 1990;10: 4180–91.
specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell 1992;71: 527–42.
noncoding RNA mediates the prometastatic functions of TGF-b. Cancer Cell 2014;25:557–9.
27. Zhao J, Sun BK, Erwin JA, et al. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 2008;322:750–6.
39. Tripathi V, Ellis JD, Shen Z, et al. The nuclearretained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell 2010;39:925–38.
28. Surani MA, Barton SC, Norris ML. Development of reconstituted mouse eggs suggests imprinting
40. Engreitz JM, Sirokman K, McDonel P, et al. RNA-RNA interactions enable specific targeting of
of the genome during gametogenesis. Nature 1984;308:548–50.
noncoding RNAs to nascent pre-mRNAs and chromatin sites. Cell 2014;159:188–99.
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
41. Faghihi MA, Zhang M, Huang J, et al. Evidence for natural antisense transcript-mediated inhibition of microRNA function. Genome Biol 2010;11: R56. 42. Giovarelli M, Bucci G, Ramos A, et al. H19 long noncoding RNA controls the mRNA decay promoting function of KSRP. Proc Natl Acad Sci U S A 2014;111:E5023–8. 43. Carrieri C, Cimatti L, Biagioli M, et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 2012;491:454–7. 44. Meurs KM, Mauceli E, Lahmers S, et al. Genome-wide association identifies a deletion in the 30 untranslated region of striatin in a canine model of arrhythmogenic right ventricular cardiomyopathy. Hum Genet 2010;128:315–24. 45. Miller CL, Haas U, Diaz R, et al. Coronary heart disease-associated variation in TCF21 disrupts a miR-224 binding site and miRNA-mediated regulation. PLoS Genet 2014;10:e1004263. 46. Helgadottir A, Thorleifsson G, Manolescu A, et al. A common variant on chromosome 9p21 affects the risk of myocardial infarction. Science 2007;316:1491–3. 47. McPherson R, Pertsemlidis A, Kavaslar N, et al. A common allele on chromosome 9 associated with coronary heart disease. Science 2007;316: 1488–91. 48. Samani NJ, Erdmann J, Hall AS, et al., for WTCCC and the Cardiogenics Consortium. Genomewide association analysis of coronary artery disease. N Engl J Med 2007;357:443–53. 49. Holdt LM, Teupser D. From genotype to phenotype in human atherosclerosis—recent findings. Curr Opin Lipidol 2013;24:410–8. 50. Erridge C, Gracey J, Braund PS, et al. The 9p21 locus does not affect risk of coronary artery disease through induction of type 1 interferons. J Am Coll Cardiol 2013;62:1376–81. 51. Holdt LM, Teupser D. Recent studies of the human chromosome 9p21 locus, which is associated with atherosclerosis in human populations. Arterioscler Thromb Vasc Biol 2012;32:196–206. 52. Holdt LM, Beutner F, Scholz M, et al. ANRIL expression is associated with atherosclerosis risk at chromosome 9p21. Arterioscler Thromb Vasc Biol 2010;30:620–7. 53. Congrains A, Kamide K, Oguro R, et al. Genetic variants at the 9p21 locus contribute to atherosclerosis through modulation of ANRIL and CDKN2A/B. Atherosclerosis 2012;220:449–55. 54. Tsai PC, Liao YC, Lin TH, et al. Additive effect of ANRIL and BRAP polymorphisms on anklebrachial index in a Taiwanese population. Circ J 2012;76:446–52. 55. Harismendy O, Notani D, Song X, et al. 9p21 DNA variants associated with coronary artery disease impair interferon-g signalling response. Nature 2011;470:264–8. 56. Holdt LM, Hoffmann S, Sass K, et al. Alu elements in ANRIL non-coding RNA at chromosome 9p21 modulate atherogenic cell functions through trans-regulation of gene networks. PLoS Genet 2013;9:e1003588.
Long Noncoding RNAs in Cardiovascular Disease
57. Burd CE, Jeck WR, Liu Y, et al. Expression of linear and novel circular forms of an INK4/ARFassociated non-coding RNA correlates with atherosclerosis risk. PLoS Genet 2010;6: e1001233. 58. Congrains A, Kamide K, Katsuya T, et al. CVDassociated non-coding RNA, ANRIL, modulates expression of atherogenic pathways in VSMC. Biochem Biophys Res Commun 2012;419:612–6. 59. Kotake Y, Nakagawa T, Kitagawa K, et al. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 2011;30:1956–62. 60. Bochenek G, Häsler R, El Mokhtari NE, et al. The large non-coding RNA ANRIL, which is associated with atherosclerosis, periodontitis and several forms of cancer, regulates ADIPOR1, VAMP3 and C11ORF10. Hum Mol Genet 2013;22: 4516–27. 61. Zhao W, Smith JA, Mao G, et al. The cis and trans effects of the risk variants of coronary artery disease in the Chr9p21 region. BMC Med Genomics 2015;8:21. 62. Ishii N, Ozaki K, Sato H, et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J Hum Genet 2006;51: 1087–99. 63. Gao W, Zhu M, Wang H, et al. Association of polymorphisms in long non-coding RNA H19 with coronary artery disease risk in a Chinese population. Mutat Res 2015;772:15–22. 64. Matouk IJ, Halle D, Gilon M, et al. The noncoding RNAs of the H19-IGF2 imprinted loci: a focus on biological roles and therapeutic potential in lung cancer. J Transl Med 2015;13:113. 65. Kim DK, Zhang L, Dzau VJ, et al. H19, a developmentally regulated gene, is reexpressed in rat vascular smooth muscle cells after injury. J Clin Invest 1994;93:355–60. 66. Han DK, Khaing ZZ, Pollock RA, et al. H19, a marker of developmental transition, is reexpressed in human atherosclerotic plaques and is regulated by the insulin family of growth factors in cultured rabbit smooth muscle cells. J Clin Invest 1996;97: 1276–85. 67. Tragante V, Barnes MR, Ganesh SK, et al. Gene-centric meta-analysis in 87,736 individuals of European ancestry identifies multiple bloodpressure-related loci. Am J Hum Genet 2014;94: 349–60. 68. Kallen AN, Zhou XB, Xu J, et al. The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell 2013;52:101–12. 69. Yan L, Zhou J, Gao Y, et al. Regulation of tumor cell migration and invasion by the H19/let-7 axis is antagonized by metformin-induced DNA methylation. Oncogene 2015;34:3076–84. 70. Bao MH, Feng X, Zhang YW, et al. Let-7 in
72. Grote P, Wittler L, Hendrix D, et al. The tissuespecific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse. Dev Cell 2013;24:206–14. 73. Kurian L, Aguirre A, Sancho-Martinez I, et al. Identification of novel long noncoding RNAs underlying vertebrate cardiovascular development. Circulation 2015;131:1278–90. 74. May D, Blow MJ, Kaplan T, et al. Large-scale discovery of enhancers from human heart tissue. Nat Genet 2011;44:89–93. 75. Blow MJ, McCulley DJ, Li Z, et al. ChIP-Seq identification of weakly conserved heart enhancers. Nat Genet 2010;42:806–10. 76. Matkovich SJ, Edwards JR, Grossenheider TC, et al. Epigenetic coordination of embryonic heart transcription by dynamically regulated long noncoding RNAs. Proc Natl Acad Sci U S A 2014;111: 12264–9. 77. Uchida S, Dimmeler S. Long non-coding RNAs in cardiovascular diseases. Circ Res 2015;116: 737–50. 78. Boeckel JN, Jaé N, Heumüller AW, et al. Identification and characterization of hypoxiaregulated endothelial circular RNA. Circ Res 2015;117:884–90. 79. Yang KC, Yamada KA, Patel AY, et al. Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation 2014;129:1009–21. 80. Ounzain S, Micheletti R, Beckmann T, et al. Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs. Eur Heart J 2015;36:353–68a. 81. Di Salvo TG, Guo Y, Su YR, et al. Right ventricular long noncoding RNA expression in human heart failure. Pulm Circ 2015;5:135–61. 82. Carrion K, Dyo J, Patel V, et al. The long noncoding HOTAIR is modulated by cyclic stretch and WNT/b-CATENIN in human aortic valve cells and is a novel repressor of calcification genes. PLoS One 2014;9:e96577. 83. Gupta RA, Shah N, Wang KC, et al. Long noncoding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010;464: 1071–6. 84. Kumarswamy R, Bauters C, Volkmann I, et al. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ Res 2014;114:1569–75. 85. Dorn GW II LIPCAR: a mitochondrial lnc in the noncoding RNA chain? Circ Res 2014;114:1548–50. 86. Vausort M, Wagner DR, Devaux Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res 2014;115:668–77.
cardiovascular diseases, heart development and cardiovascular differentiation from stem cells. Int J Mol Sci 2013;14:23086–102.
87. Yang Y, Cai Y, Wu G, et al. Plasma long noncoding RNA, CoroMarker, a novel biomarker for diagnosis of coronary artery disease. Clin Sci (Lond) 2015;129:675–85.
71. Klattenhoff CA, Scheuermann JC, Surface LE,
88. Lorenzen JM, Schauerte C, Kielstein JT, et al.
et al. Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell 2013; 152:570–83.
Circulating long noncoding RNATapSaki is a predictor of mortality in critically ill patients with acute kidney injury. Clin Chem 2015;61:191–201.
1225
1226
Boon et al.
JACC VOL. 67, NO. 10, 2016 MARCH 15, 2016:1214–26
Long Noncoding RNAs in Cardiovascular Disease
89. Memczak S, Papavasileiou P, Peters O, et al. Identification and characterization of circular RNAs as a new class of putative biomarkers in human blood. PLoS One 2015;10:e0141214.
99. Fiedler J, Breckwoldt K, Remmele CW, et al. Development of long noncoding RNA-based strategies to modulate tissue vascularization. J Am Coll Cardiol 2015;66:2005–15.
90. Boeckel JN, Thomé CE, Leistner D, et al.
100. Wu G, Cai J, Han Y, et al. LincRNA-p21 reg-
Heparin selectively affects the quantification of microRNAs in human blood samples. Clin Chem 2013;59:1125–7.
ulates neointima formation, vascular smooth muscle cell proliferation, apoptosis, and atherosclerosis by enhancing p53 activity. Circulation 2014;130:1452–65.
91. Bassett AR, Akhtar A, Barlow DP, et al. Considerations when investigating lncRNA function in vivo. Elife 2014;3:e03058. 92. Maeder ML, Linder SJ, Cascio VM, et al. CRISPR RNA-guided activation of endogenous human genes. Nat Methods 2013;10:977–9. 93. Michalik KM, You X, Manavski Y, et al. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ Res 2014;114:1389–97. 94. Yan B, Tao ZF, Li XM, et al. Aberrant expression of long noncoding RNAs in early diabetic retinopathy. Invest Ophthalmol Vis Sci 2014;55:941–51. 95. Liu JY, Yao J, Li XM, et al. Pathogenic role of lncRNA-MALAT1 in endothelial cell dysfunction in diabetes mellitus. Cell Death Dis 2014;5:e1506. 96. Puthanveetil P, Chen S, Feng B, et al. Long non-coding RNA MALAT1 regulates hyperglycaemia induced inflammatory process in the endothelial cells. J Cell Mol Med 2015;19:1418–25. 97. Jaé N, Dimmeler S. Long noncoding RNAs in diabetic retinopathy. Circ Res 2015;116:1104–6. 98. Yan B, Yao J, Liu JY, et al. lncRNA-MIAT regulates microvascular dysfunction by functioning as a competing endogenous RNA. Circ Res 2015;116: 1143–56.
101. Hu YW, Zhao JY, Li SF, et al. RP5–833A20.1/ miR-382–5p/NFIA-dependent signal transduction pathway contributes to the regulation of cholesterol homeostasis and inflammatory reaction. Arterioscler Thromb Vasc Biol 2015;35:87–101. 102. Bell RD, Long X, Lin M, et al. Identification and initial functional characterization of a human vascular cell-enriched long noncoding RNA. Arterioscler Thromb Vasc Biol 2014;34:1249–59. 103. Li P, Ruan X, Yang L, et al. A liver-enriched long non-coding RNA, lncLSTR, regulates systemic lipid metabolism in mice. Cell Metab 2015;21:
107. Han P, Li W, Lin CH, et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature 2014;514:102–6. 108. Hinkel R, Penzkofer D, Zühlke S, et al. Inhibition of microRNA-92a protects against ischemia/ reperfusion injury in a large-animal model. Circulation 2013;128:1066–75. 109. Janssen HLA, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med 2013;368:1685–94. 110. Krieg AM. Targeting LDL cholesterol with LNA. Mol Ther Nucleic Acids 2012;1:e6. 111. Jeong W, Rapisarda A, Park SR, et al. Pilot trial of EZN-2968, an antisense oligonucleotide inhibitor of hypoxia-inducible factor-1 alpha (HIF-1a), in patients with refractory solid tumors. Cancer Chemother Pharmacol 2014;73:343–8. 112. Tanioka M, Nokihara H, Yamamoto N, et al. Phase I study of LY2181308, an antisense oligo-
455–67.
nucleotide against survivin, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2011;68:505–11.
104. Wang K, Sun T, Li N, et al. MDRL lncRNA regulates the processing of miR-484 primary transcript by targeting miR-361. PLoS Genet 2014; 10:e1004467.
113. Denzler R, Agarwal V, Stefano J, et al. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol Cell 2014;54:766–76.
105. Wang K, Liu F, Zhou LY, et al. The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489. Circ Res 2014;114: 1377–88. 106. Wang K, Liu CY, Zhou LY, et al. APF lncRNA regulates autophagy and myocardial infarction by targeting miR-188–3p. Nat Commun 2015;6:6779.
114. Anderson DM, Anderson KM, Chang CL, et al. A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell 2015;160:595–606.
KEY WORDS cardiac, long noncoding RNA, microRNA, vascular