You are on page 1of 10

M I N I R E V I E W

MicroRNAs in Cardiovascular Health: From Order to


Disorder
Denuja Karunakaran and Katey J. Rayner
University of Ottawa Heart Institute, Ottawa, Ontario K1Y4W7 Canada

In the last decade, microRNAs (miRNAs) have revolutionized how we understand metabolism and
disease. These small, 20- to 22-nucleotide RNA molecules fine-tune gene expression and can often
coordinate multiple genes in a single pathway. Given the multifactorial nature of cardiovascular dis-
ease, it is perhaps not surprising that miRNAs have been shown to orchestrate many aspects of disease
development, from modulating metabolic risk factors over a lifetime (eg, cholesterol and hormones)
to controlling the response to an acute cardiovascular event (eg, inflammation and hypoxia). In this
review, we discuss how miRNAs exert control over metabolic pathways that maintain vascular health
and, when these pathways go awry, how miRNAs can be targeted for therapeutic modulation.
(Endocrinology 154: 4000 4009, 2013)

n the three decades since the discovery and subsequent lar health and, when these pathways go awry, how miRNAs
I widespread use of statins as lipid-lowering agents, there
has been a paucity of new therapies to treat and manage
can be targeted for therapeutic modulation.

the risk factors for cardiovascular disease (1). Although


sudden death from cardiac events (ie, myocardial infarc- The miRNA Revolution
tion [MI]) has decreased dramatically over the past 20
years, a significant majority of the population in the west- The miRNAs were first discovered in 1993, when Victor
ern world continues to suffer from hyperlipidemia, over- Ambros and colleagues (3) undertook a genetic screen to
nutrition, and obesity, all risk factors for cardiometabolic identify essential genes for lineage commitment in developing
disease (2). In the past, drug development has largely fo- Caenorhabditis elegans. They unexpectedly found that a
cused on pharmacological inhibition of single targets, of- small antisense RNA (termed lin-4) was binding to and in-
ten using small molecules, as a means to modulate bio- hibiting the expression of a protein-coding transcript, LIN-
logical pathways that promote cardiovascular disease 14, which was required for commitment to the first postem-
and/or its associated complications. However, this ap- bryonic larval stage. Nearly a decade later, it was revealed
proach has been plagued with difficulties, owing largely to that antisense RNA was not just an anomaly in worms but
the lack of tissue specificity of small molecules as well as that it was highly conserved and even found in humans (4).
their often dangerous off-target effects. Hence, there Almost immediately, additional miRNAs were discovered in
remains an urgent need to refocus efforts in therapeutic both C. elegans and mammals, and so began the miRNA
development away from pharmacologic modulators revolution (57). To date, there have been over a thousand
and instead exploit newly uncovered molecules and mech- individual miRNAs annotated in the human genome, with
anisms. MicroRNAs (miRNAs) are now understood to be more added every year, and over 50% of the human genome
powerful modulators of biological pathways and have is estimated to be under miRNA control (8, 9).
generated considerable excitement as next-generation miRNAs begin as long, primary transcripts that are
therapy for a variety of diseases, including cardiometa- most commonly transcribed by RNA polymerase II from
bolic disease. In this review, we will discuss how miRNAs intergenic, intronic, or even exonic locations in the ge-
exert control over metabolic pathways that maintain vascu- nome (10). They are cleaved in the nucleus by the ribo-

ISSN Print 0013-7227 ISSN Online 1945-7170 Abbreviations: ABC, ATP-binding cassette transporters; EC, endothelial cell; eNOS, endo-
Printed in U.S.A. thelial NO synthase; ER, estrogen receptor-; HDL, high-density lipoprotein; KLF4, Krp-
Copyright 2013 by The Endocrine Society pel-like factor 4; LDL, low-density lipoprotein; MI, myocardial infarction; miRNA, mi-
Received March 31, 2013. Accepted August 26, 2013. croRNA; NFB, nuclear factor kappa-light-chain-enhancer of activated B cells; PI3K,
First Published Online September 5, 2013 phosphoinositide 3-kinase; pre-miRNA, precursor miRNA; SMC, smooth muscle cell;
VCAM-1, vascular cell adhesion molecule 1.

4000 endo.endojournals.org Endocrinology, November 2013, 154(11):4000 4009 doi: 10.1210/en.2013-1299

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
doi: 10.1210/en.2013-1299 endo.endojournals.org 4001

nuclease III enzyme-complex Drosha/DGCR8 (DiGeorge LDL deposits in the subendothelial space, where it is sub-
Syndrome chromosomal region 8, also known as pasha) to jected to oxidative damage by the environment rich in reac-
form smaller precursor miRNA (pre-miRNA) products. This tive oxygen species and oxidizing enzymes like myeloperox-
microprocessor complex is exported from the nucleus where idase (16). Macrophages and other inflammatory cells
it is further cleaved by Dicer to form the mature, 20- to 22- present in atherosclerotic plaques are an abundant source of
nucleotide double-stranded miRNA molecules. One strand, both reactive oxygen species and myeloperoxidase, and their
termed the effector/functional strand, of the miRNA is persistence in lesions causes an accumulation of these oxi-
loaded into the RNA-induced silencing complex, which con- dized LDL products. The uptake of oxidized LDL through
sists of RNA binding proteins like the Argonautes (ie, Ago1 scavenger receptor CD36 is itself inflammatory, which
and Ago2) with cleavage activity. The RNA-induced silenc- causes additional recruitment and retention of macrophages
ing complex is guided to complementary regions in specific to the lesional area, propagating the inflammatory cascade
mRNA transcripts, most often (but not exclusively) located and ultimately lesion progression (17). Excess lipid accumu-
in 3-untranslated regions of target genes. Until recently, it lates in macrophage foam cells and, if not efficiently removed
was believed that the passenger strand (previously denoted through the cholesterol efflux pathway, largely mediated by
with an asterisk) was simply degraded and was thus disre- the cholesterol transporters ATP-binding cassette transport-
garded as nonfunctional; however, evidence is mounting that ers (ABC), ABCA1 and ABCG1, will lead to additional in-
these passenger strands can also be functional and bind flammatory activation and even cell death. On the other
mRNA transcripts under certain circumstances (11). Com- hand, HDL acts as an opposing force to LDL and can enter
plete complementary base pairing between the miRNA:m- the subendothelial space and act as an acceptor for the excess
RNA duplex results in destabilization or degradation of the cholesterol that is effluxed through ABCA1 and ABCG1.
mRNA, whereas partial base pairing results in deadenylation This cholesterol is then brought to the liver, taken up via
or translational inhibition (12, 13). Generally speaking, the scavenger receptor BI, and excreted through the bile and feces
consequence of miRNA:mRNA duplex formation is a down- in a process known as reverse cholesterol transport. HDL is
regulation of target protein expression, although exceptions made by the liver, intestine, and adipose tissue by ABCA1, yet
to this have been reported (14, 15). Frequently, the magni- it is the efficiency of reverse cholesterol transport that is be-
tude of protein repression enforced by miRNA is modest; lieved to underlie the benefits often ascribed to high levels of
that is, any one miRNA may cause only a 20% decrease in HDL and is now being touted as the most important metric
individual target gene protein expression under basal condi- of HDL function (18).
tions. However, miRNAs often repress multiple members of miRNAs have been shown to control lipoproteins and
the same pathway simultaneously, making their cumulative their pro-/antiatherogenic effects at many levels (Figure
impact on cellular function more potent and dramatic. Given 1A). The first miRNA demonstrated to specifically mod-
the multifactorial nature of cardiovascular disease, it is per- ulate cholesterol balance was miR-33. miR-33 is a lipid-
haps not surprising that miRNAs have been shown to or- responsive miRNA embedded in the introns of the sterol
chestrate many aspects of disease development, from mod- regulatory transcription factor genes SREBP-1 and
ulating metabolic risk factors over a lifetime (eg, cholesterol SREBP-2 and is cotranscribed along with its host genes
and hormones) to controlling the response to an acute car- under conditions that induce SREBP activation (19 21).
diovascular event (eg, inflammation and hypoxia). miR-33 represses the expression of ABCA1, ABCG1, and
NPC1, which ultimately prevents cholesterol export from
cells. Inhibition of miR-33 was shown to induce the ex-
Metabolic Risk Factor Regulation by pression of ABCA1 not only in the liver, where it increased
miRNAs circulating HDL levels but also in atherosclerotic plaque
macrophages where it promoted cholesterol removal from
Regulation of cholesterol and lipoproteins by miRNAs the plaque and decreased lesion size (22). Genetic deletion
Lipids are carried in the bloodstream by lipoproteins of miR-33 also resulted in sustained increases in HDL as
where they are shuttled to and from tissues as required. Low- well as a decrease in atherosclerosis progression in
density lipoprotein (LDL) delivers cholesterol and phospho- Apoe/ mice (23, 24). miR-33 can also control the fatty
lipids from the liver to tissues in need, whereas high-density acid oxidation and synthesis pathway, and modulation of
lipoprotein (HDL) ferries excess lipids away from peripheral miR-33 levels in nonhuman primates reduced circulating
tissues back to the liver for excretion. High levels of LDL very-low-density lipoprotein, another important risk fac-
cholesterol and low levels of HDL cholesterol are both in- tor for cardiometabolic disease (25). Another miRNA,
dependent risk factors for the development of atherosclerosis miR-144, was recently shown to modulate ABCA1 ex-
and its downstream complications. Consequently, excess pression and cholesterol efflux from hepatocytes, and in-

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
4002 Karunakaran and Rayner miRNA and Cardiovascular Health Endocrinology, November 2013, 154(11):4000 4009

hibition of miR-144 in vivo increases hepatic ABCA1 and Estrogen and other hormones are under the
circulating HDL (26, 27). Unlike that of HDL, the influence of miRNAs
miRNA-mediated control of LDL cholesterol has been less Steroid hormones are an essential component of develop-
well studied. miR-122, the most abundant miRNA ex- ment and are required for homeostatic function of many
pressed in liver, controls both LDL and HDL cholesterol tissues, including the vessel wall. The importance of estrogen
levels, primarily by indirect modulation of cholesterol syn- and its cognate receptors, estrogen receptor- (ER) and
thesis pathways, although the exact mechanisms remain in- ER, on both the physiology and pathophysiology of the
completely understood (28 30). Recently, miR-27b was cardiovascular system have been well described (3234). In
identified as a potential regulatory hub of lipid metabolism in brief, estrogen promotes vasodilation through induction of
the liver, potentially controlling both HDL cholesterol and endothelial NO synthase (eNOS) by vascular endothelial
triglyceride levels (31). It is interesting to postulate that miR- cells (ECs), while it also prevents vascular smooth muscle cell
27b could be exerting control over an entire network of lipid (SMC) proliferation and contraction (35, 36). Moreover, es-
metabolism genes, although these observations have yet to be trogen has been shown to reduce cytokine expression in the
systematically validated in vivo. vessel wall (37) and has even been demonstrated to reduce

Figure 1. microRNAs in cardiovascular health. A, miRNAs controls lipid metabolism, lipoprotein clearance and their pro- or anti-atherogenic
effects at multiple sites. B, In the vessel wall, estrogen regulates miRNA function in vascular cells. C, miRNAs target multiple cell types and
functions within the vessel wall to modulate inflammatory responses and atherosclerosis progression. D, Several miRNAs have been shown to
intricately regulate cardiac fibrosis, hypertrophy and remodelling and repair post-ischemic injury or MI.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
doi: 10.1210/en.2013-1299 endo.endojournals.org 4003

LDL and raise HDL through modulation of apolipoprotein (nuclear factor kappa-light-chain-enhancer of activated B
expression (32). These beneficial effects of estrogen underlie cells) and regulator of vesicular trafficking of insulin, and
the observation that premenopausal women, by and large, 3-phosphoinositide-dependent protein kinase-1 (Pdk1), an
have lower rates of cardiovascular disease than postmeno- integral constituent of the insulin-mediated PI3K-AKT/PKB
pausal women, and postmenopausal women are at greater (phosphoinositide 3-kinase-AKT/protein kinase B) signaling
risk of dying from heart disease than age-matched men (38). pathways (42, 43). miR-375 knockout mice have decreased
Although clinical trials have been confusing and often -cell mass and increased gluconeogenesis and
fraught with controversy, most of the evidence supports the hepatic glucose output and are hyperglycemic, demonstrat-
notion that hormone replacement therapy immediately after ing the important role miR-375 has in regulating glucose
the onset of menopause protects against cardiovascular dis- homeostasis (43). Interestingly, both diabetic patients (44)
ease and its associated complications. Interestingly, estrogen and ob/ob diabetic mice (43) have elevated miR-375 expres-
has been shown to control specific miRNA expression in vivo sion and -cell mass but no significant changes in -cell mass,
and in vitro, and it is believed that ER is primarily respon- suggesting miR-375 overexpression may contribute to the
sible for these effects (39). This is accomplished through di- observed decreased insulin and the resultant high blood glu-
rect interaction of ER with the microprocessor complex cose levels, all contributing factors for type 2 diabetes. Ago2
Drosha, which governs the cleavage of primary miRNA tran- has recently been implicated in regulating pancreatic -cell
scripts to the pre-miRNA transcripts (39). Notably, ER- exocytosis, because small interfering RNA knockdown of
mediated destabilization of Drosha decreased the expression Ago2 increased expression of miR-375 target genes gephyrin
of miR-125a and miR-16, which then provided a positive- and ywhaz, which enhanced insulin secretion in response to
feedback loop through derepression of ER-induced target glucose (45). Thus, miRNA-mediated control of pancreatic
genes (39). This elegant study revealed the complexity of gene expression is essential for proper insulin secretion and
estrogen and ER in the biogenesis of miRNA and began to homeostasis. Pancreatic -cell function is regulated in a dis-
shed light on the multifaceted nature of estrogen action on tinct manner by miR-33. Cholesterol homeostasis in pancre-
cellular function (Figure 1B). In the vessel wall, estrogen con- atic -cells is maintained by ABCA1, and regulation of cel-
trols the expression of miR-203, which then negatively reg- lular cholesterol content is required for proper insulin
ulates the cellular proliferation program through Abl1 and secretion (46). Similar to macrophages and hepatocytes, as
p63 (40). Although these observations have yet to be con- mentioned above, overexpression of miR-33 also inversely
firmed in vivo, this was the first study demonstrating a role modulates ABCA1 expression in pancreatic -cells, and this
for estrogen-mediated miRNA expression in controlling vas- in turn corresponds to higher cholesterol levels and decreased
cular cell homeostasis, and full examination of the spectrum glucose-mediated insulin exocytosis from these cells (47). In-
of miRNA-mediated estrogen action in the vasculature is hibition of miR-33 improved insulin secretion from islet cells
anticipated. in vitro from hypercholesterolemic mice, suggesting that this
pathway is indeed impaired in these mice. miR-33 also mod-
Control of obesity, insulin signaling, and glucose ulates the tissue response to insulin by its regulation of insulin
regulation by miRNAs receptor substrate 2 (IRS2), a critical component in mediat-
Type 2 diabetes is a chronic metabolic disorder caused by ing insulin-signaling pathways, and, when impaired, con-
a combination of altered insulin secretion and progressive tributes to insulin resistance (48). Although the effects of
tissue insulin resistance. Diabetic complications can further miR33 inhibition therapy have yet to be tested in models of
exacerbate cardiovascular risk factors such as hypertension, insulin resistance or diabetes in vivo, it would stand to reason
dyslipidemia, obesity, and heart failure, all of which increase that antimiR-33 would promote glucose-mediated insulin
the overall cardiovascular disease burden (2). In healthy in- exocytosis from -cells while simultaneously restoring insu-
dividuals, high blood glucose levels stimulate pancreatic lin-mediated signaling pathways in peripheral tissues and,
-cells to secrete insulin, promoting the uptake of glucose by thus, improving the development of atherosclerosis and
peripheral target tissues such as skeletal muscle and adipose diabetes.
tissue (41). Glucose homeostasis is stringently regulated by a In addition to its release from pancreatic -cells, miRNAs
variety of interacting signaling pathways that synchronize modulate the downstream response to insulin by altering
both gene and protein expression and function, and miRNAs components of the insulin signaling machinery in periph-
were recently identified as another level of posttranscrip- eral tissues. miR-103 and miR-107, which differ by only
tional regulation of these pathways (Figure 1A). The most one nucleotide, are necessary for insulin signaling through
abundant miRNA in pancreatic endocrine cells, miR-375, the insulin receptor (49). Inactivation or silencing of miR-
negatively regulates glucose-induced insulin secretion by di- 103/107 upregulates caveolin-1, which in turn stabilizes
rectly repressing myotrophin (MTPN), an activator of NFB the insulin receptor and promotes insulin sensitivity in the

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
4004 Karunakaran and Rayner miRNA and Cardiovascular Health Endocrinology, November 2013, 154(11):4000 4009

liver and adipose tissue. Notably, the expression of miR- marrow into high-fat diet-fed LDLR/ mice resulted in in-
103/107 is raised in the liver of both obese and type 2 creased atherosclerotic lesion development and increased le-
diabetic mouse models (49, 50), and inactivation of these sional inflammation (55, 56). However, hyperlipidemic
miRNAs improves glucose homoeostasis in diabetic mice apoE/ mice transplanted with miR-155/ bone marrow
(49). miR-143/145 expression is similarly upregulated in displayed decreased macrophage content and reduced ath-
the liver of obese and diabetic mice (51). miR-143 and erosclerotic plaque size (55). In macrophage foam cells, miR-
miR-145 target the AKT signaling pathway, and mice de- 155 targets Bcl6, an NFB antagonist, which promotes pro-
ficient in the miR-143/145 cluster are protected from the inflammatory signals such as CCL2 expression (55).
development of diet-induced insulin resistance. Taken to- However, miR-155 is also required for early cell fate deci-
gether, these studies suggest that manipulating miRNA sions in inflammatory cells, and deficiency of miR-155 alters
clusters (rather than a single miRNA) could present po- the profile of monocytes and neutrophils in miR-155/ hy-
tential therapies to treat obesity and the development of perlipidemic mice (56). The role of miR-155 in inflammatory
type 2 diabetes. precursor cell fate vs advanced macrophage foam cells likely
underlies the discrepancy observed between the two mouse
models and highlights the dual role of miR-155 in promoting
Control of Cardiovascular Inflammation atherosclerosis. Interestingly, miR-3425p was found to di-
and Injury by miRNAs rectly activate the expression of miR-155 and is upregulated
in early atherosclerotic lesions of apoE/ mice (57). This in
Inflammation in the vessel wall is regulated by
turn induces proinflammatory mediators such as Nos2 and
multiple miRNAs
IL-6. Furthermore, miR-155 expression is upregulated in
As described above, modified lipoproteins trapped in the
ECs and appears to suppress angiotensin II type I receptor
vessel wall instigate the inflammatory response that underlies
expression in these cells, reducing the proinflammatory and
atherosclerosis and the dangerous development of the un-
vascular remodeling roles of angiotensin II (59). The thera-
stable, rupture-prone plaque. Apart from monocytes/mac-
peutic potential of miR-155 will thus depend on the stage of
rophages, other key cells involved in this inflammatory cas-
disease when therapy is initiated but will likely have benefi-
cade include ECs, vascular SMCs, leukocytes, and platelets.
cial outcomes in the patient with advanced atherosclerotic
ECs are considered the inflammatory gatekeepers of the ves-
sel wall, and resting ECs do not express inflammatory mark- disease.
ers like VCAM-1 (vascular cell adhesion molecule-1). Upon Encompassed in the same gene cluster but with little ho-
activation by cytokines, they increase their expression of mology to each other, both miR-143 and miR-145 are highly
VCAM-1 and recruit inflammatory cells to the subendothe- conserved and expressed in SMCs, and their expression is
lial space, and it is the constant recruitment of these cells into markedly reduced in coronary artery disease patients (54, 60,
the intima that leads to the formation of large atherosclerotic 61). Apart from their role in the pancreas, miR-143/145 ex-
lesions. miR-126 in ECs directly targets the untranslated re- pression is also upregulated by transcription factors in vessel
gion of VCAM-1 to repress its expression, and inhibition of wall SMCs, resulting in a combinatorial effect where miR-
miR-126 upregulates VCAM-1 expression and leukocyte 143 inhibits SMC proliferation and miR-145 promotes SMC
adherence to ECs (52). Interestingly, EC apoptotic bodies differentiation through direct targeting of Krppel-like fac-
shed microvesicles containing miR-126, which represses tor 4 (KLF4), myocardin, and Elk-1 (ELK1) as well as an-
RGS16 (regulator of G protein signaling 16) in arteries, de- giotensin converting enzyme (ACE) and its cognate receptor
creases production of the inflammatory chemokine (60, 61). Genetic knockdown of miR-143/145 results in the
CXCL12 (chemokine (C-X-C motif) ligand 12), decreases development of spontaneous atherosclerotic lesions in old
recruitment of inflammatory cells, and overall reduces in- mice (60). SMC-specific overexpression of miR-145 using
flammation and stabilizes the atherosclerotic plaque (53). lentivirus in mice resulted in reduced plaque size, reduced
miR-126 is now recognized as an important regulator of necrotic core, increased fibrous cap area, and increased
vascular EC maintenance and is integral to the progression of plaque collagen content, all of which may contribute to sta-
atherosclerosis (Figure 1C). bilizing the plaque (62). Additionally, the vascular shear-
miR-155 was recently found to be upregulated in ath- responsive transcription factor KLF2 upregulates the miR-
erosclerotic lesions in both mice and humans and is decreased 143/145 cluster in ECs, and these miRs are secreted into the
in the circulation of coronary artery disease patients (54, 55). extracellular space, where they are thought to target SMCs
Despite these observations, the data supporting the exact role and thus reduce atherosclerosis lesion progression in
of miR-155 in the progression of atherosclerosis is contra- apoE/ mice (63). Thus, miR-143/145 regulates vascular
dictory to date. Transplantation of miR-155/ mice bone wall homeostasis from both intracellular and extracellular

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
doi: 10.1210/en.2013-1299 endo.endojournals.org 4005

mechanisms, adding another layer of complexity in ancies between these two studies could be attributable to the
miRNA-mediated gene regulation. anti-miR therapies used, which differ in their pharmacoki-
Exaggerated platelet responses or hyperactivity is thought netic profiles, especially with respect to potency, toxicity,
to contribute to the development of thrombotic and occlu- tissue uptake, and off-target effects. The observation that
sive events that lead to life-threatening MI. Because platelets cardiac remodeling was unaffected with long-term repres-
are anucleated and possess no genomic DNA and little sion of miR-21, such as in the knockout model, suggests that
mRNA, there had been minimal focus on understanding there may be miRNA redundancy or compensatory mecha-
transcriptional regulation in platelet function until recently. nisms at play. Additionally, inhibition of miR-21 in SMCs
Landry et al (64) performed a comprehensive study of miR- upregulates phosphatase and tensin homolog (PTEN),
NAs in platelets, demonstrating the presence of 219 miRNAs which in turn inhibits the PI3K/AKT pathways resulting in
and Dicer and Ago2 complexes; however, understanding the decreased SMC proliferation and increased apoptosis, caus-
role of platelet miRNAs is in its infancy. miR-223 has been ing attenuation of neointima formation after angioplasty
implicated as a key regulator of P2Y12 (purinergic receptor (71). Caution must be taken in considering miR-21targeted
P2Y, G-protein coupled 12), which binds to soluble ADP therapies for cardiovascular disease, due to the ubiquitous
that is released from activated platelet granules to further expression of miR-21, which makes the development of tis-
propagate the platelet aggregation response (64). Another sue- or cell-specific therapies essential.
study further confirmed a functional role for platelet miR- Expressed solely in the heart, miR-208a has been shown
NAs, and here they implicated that miR-200b can repress to regulate cardiac hypertrophy by repressing negative reg-
PRKAR2B (protein kinase cAMP-dependent type II-beta), ulators (eg, thyroid hormone-associated protein 1 and myo-
decreasing platelet responsiveness (65). It is possible that statin) of muscle growth and hypertrophy while failing to
platelet miRNAs not only serve as transcriptional regulators upregulate myosin heavy chain (MYH7), a key marker in
within platelets but also secrete miRNAs into the circulation cardiac remodeling, (72, 73). Therapeutic inhibition of this
to potentially target specific cells in response to vascular in- miRNA prevented the development of heart failure in rats via
jury (66). As a result, platelet-derived miRNAs may serve as the prevention of myosin switching in damaged hearts (74).
diagnostic biomarkers of platelet activation or post-anti- Interestingly, antimiR-208a also decreased circulating
platelet therapy (65, 67). miRNA levels in the serum, including miR-499, a biomarker
of heart injury. These data suggest that the beneficial effects
Cardiac injury and remodeling after MI: miRNAs of miR-208a inhibition may be a synergistic effect of both
play a key role myosin upregulation and decreasing circulating miRNA. In
The build-up and subsequent rupture of a vulnerable ath- addition, miR-208a regulates systemic energy homeostasis
erosclerotic plaque leads to an MI, resulting in cardiac tissue by negatively regulating MED13, and pharmacological in-
ischemia and necrosis. After restoration of blood flow, the hibition of miR-208a confers protection to diet-induced obe-
response to injury involves deposition of extracellular matrix sity by increasing MED13 expression and increasing energy
and the formation of a fibrotic scar in the damaged area. In expenditure (75). The inhibition of miR-208a thus provides
addition, inflammatory cells are recruited to the site of injury, a provocative strategy for both the repair of cardiac tissue
and angiogenesis is activated to restore blood flow to dam- after MI while simultaneously improving metabolic energy
aged tissue. Several miRNAs have been shown to intricately output to decrease overall cardiometabolic risk. Other miR-
regulate cardiac fibrosis, hypertrophy, and remodeling and NAs that are downregulated during cardiac hypertrophy in
repair post-ischemic injury or MI (Figure 1D). Perhaps the both mice and human include miR-1 and miR-133 (76).
most well-studied miRNA in the post-MI environment is Knockout of both miR-133a isoforms (miR-133a1 and miR-
miR-21, whose expression is induced in a number of path- 133a2) resulted in embryonic or postnatal lethality after se-
ological conditions (68). The role of miR-21 in cardiac re- vere cardiac fibrosis, heart failure, and cardiac malforma-
modeling is controversial. Cholesterol-modified antisense tions (77). In contrast, transgenic overexpression of miR-
oligonucleotides against miR-21 inhibited ERK-MAPK ki- 133a protected hearts from fibrosis after aortic constriction
nase pathways in cardiac fibroblasts through miR-21s spe- (78). These studies support the notion that forced expression
cific target, Spry1, resulting in the attenuation of interstitial of miR-133 may be a viable therapeutic option to reduce
fibrosis and cardiac hypertrophy and dysfunction in mice cardiac hypertrophy. Interestingly, and excitingly, a recent
(69). In contrast, another study used locked nucleic acid- study demonstrated that antagonism of miR-133 induces the
modified anti-miR oligonucleotides in vivo to inhibit miR-21 phenotypic switching of skeletal muscle progenitor cells to
and demonstrated no difference in the pathological cardiac brown adipocytes, which increases whole-body thermogen-
remodeling in response to cardiac stress, which was further esis and impedes the development of obesity (79). Thus, over-
confirmed in a miR-21 knockout model (70). The discrep- expressing miR-133 and/or inhibiting miR-208a would

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
4006 Karunakaran and Rayner miRNA and Cardiovascular Health Endocrinology, November 2013, 154(11):4000 4009

likely have benefits in cardiac tissue as well as in other tissues, advanced miRNA-targeted therapeutic, primarily used in
decreasing other cardiometabolic risk factors such as obesity. patients suffering from hepatitis C infection. This is not re-
Damage to the myocardium by MI or ischemic injury lated to the control of lipid metabolism genes by miR-122 but
requires remodeling and healing processes, which includes instead is attributable to the hijacking of host hepatic miR-
the generation of new blood vessels via angiogenesis. Both 122 for efficient viral replication and the relatively liver-spe-
ECs and SMCs are involved in these processes, and several cific expression of miR-122 (29). Overall, the safety profile
miRNAs have been implicated in regulating them. Inhibition and efficacy of anti-miR122 (known as miraversen) has been
of miR-92a in mice, a component of the highly expressed positive, with phase IIa results indicating that almost 50% of
miR-1792 cluster, increased invading ECs and formation of patients treated with anti-miR122 had undetectable hepatitis
new blood vessels through their targets integrin-5 and C viral loads after treatment (86). Although the future of
eNOS (80). Furthermore, systemic administration of the
anti-miR122 for the treatment of hepatitis C infection looks
miR-92a antagomir in an MI mouse model enhanced neo-
promising, its use for the treatment of hyperlipidemia awaits
vascularization, functional recovery of damaged tissue, in-
further study. On the other hand, anti-miR33 is another
creased left ventricular function, decreased cardiomyocyte
promising miRNA-targeting therapeutic for cardiometa-
apoptosis, and reduced infarct size (80). More recently, miR-
bolic disease, which is quickly advancing through preclinical
92a knockdown has been shown to enhance KLF4 and KLF2
expression, which is thought to be protective against athero- testing for patients with atherosclerosis, and phase I trials are
genesis (81). As indicated above, miR-126 not only increases projected to begin sometime in 2015 (84).
VCAM1 expression in ECs but also has been implicated in
maintaining vascular integrity and angiogenesis during de-
velopment through regulation of its targets, SPRED1 and Summary and Future Directions
PIK3RS (82, 83). Many other miRNAs have been implicated
The observation that dysregulated miRNA expression
in the regulation of angiogenesis (see Table 1), but many of
contributes on many levels to the pathogenesis of cardio-
them have not been studied in the context of cardiovascular
vascular disease has led to considerable excitement for
injury and repair.
their use as therapeutic targets. One could envision a pre-
ventative therapy that combines targeting of miR-33 and
miR-103/107 to improve lipid profiles and insulin resis-
Therapy for Cardiovascular Disease Based
on miRNAs tance while simultaneously targeting miR-126 and miR-
155 to reduce inflammation in the vessel wall. Or perhaps
Only a decade has elapsed since the discovery of miRNAs a more acute delivery of anti-miR-208/21 and miR-133
in humans, yet already a number of miRNA-based therapies mimics either systemically or locally after a myocardial
are gaining strides as potential therapeutics. This can be at- event could be used to improve the response to injury and
tributed to the relatively rapid development of effective, po- subsequent recovery. Although many of these miRNAs
tent anti-miRNA targeting strategies that were quickly used likely will not enter clinical trials for years to come, the
in vivo in preclinical models. Effective inhibition of miRNAs realization that miRNAs represent such an untapped
can be achieved through single-stranded antisense oligonu- source of therapeutic development has invigorated both
cleotides, similar to those used for small interfering RNA
the academic and pharmaceutical communities alike, and
silencing of mRNA gene transcripts. Various chemical mod-
the therapeutic potential of these tiny RNAs is indeed
ifications have been used to stabilize these anti-miRNA mol-
enormous.
ecules, improve their toxicity, and improve their tissue up-
take, reducing the effective dose for in vivo use (84). A
landmark study in 2005 by Krtzfeldt et al (30) used antago-
mirs (cholesterol-modified miRNA antisense molecules) in Acknowledgments
mice to demonstrate that miR-122 could be effectively si- Address all correspondence and requests for reprints to: Katey
lenced in vivo, revealing the importance of this miRNA-reg- Rayner, PhD, Assistant Professor, University of Ottawa Heart
ulated pathway in lipid metabolism. Elmn et al (85) quickly InstituteBiochemistry, 40 Ruskin Street, H4211, Ottawa, On-
followed with miR-122 inhibition studies in African green tario K1Y4W7, Canada. E-mail: krayner@ottawaheart.ca.
monkeys, which both confirmed the role of miR-122 in reg- This study was funded by CIHR Operating Grant MOP
ulating lipid metabolism in vivo as well as highlighted the 130365 (to K.J.R.) and UOHI Cardiovascular Genetics Research
therapeutic targetability of miRNAs in a model highly re- Fellowship (to D.K.).
lated to humans. To date, anti-miR122 remains the most Disclosure Summary: The authors have nothing to disclose.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
doi: 10.1210/en.2013-1299 endo.endojournals.org 4007

Table 1. miRNAs Regulating Cardiometabolic Health/Disease


Risk Factors and
miRNAs Target Cell/Tissue Effect Ref.
Cholesterol and
lipoproteins
miR-33 ABCA1, ABCG1, NPC1 Macrophages, liver HDL/cholesterol efflux 19 21
miR-144 ABCA1 Liver HDL/cholesterol efflux 26, 27
miR-122 ? (cholesterol synthesis pathways) Liver LDL, HDL 28 30
miR-27b Angptl3, Gpam Hepatocytes HDL, triglycerides 31
Hormones (eg, estrogen)
miR-16, miR-125a ER gene targets Breast cancer Estrogen-responsive gene changes 39
miR-203a Abl1, p63 Vascular SMCs SMC proliferation 40
Insulin resistance and
obesity
miR-375 NFB, PDK1 Pancreatic endocrine cells Insulin vesicular trafficking and 42, 43
exocytosis, - and -cell mass
Ago2 Gephyrin, ywhaz Pancreatic cell line, MIN6 Insulin exocytosis 45
miR-33 ABCA1, IRS2 Islet cells Cholesterol efflux, insulin 47, 48
exocytosis
miR-103, miR-107 Caveolin-1 Peripheral tissues (eg, liver, Stabilization of insulin receptor, 49, 50
adipose) insulin sensitivity
miR-143/145 AKT liver Insulin receptor signaling? 51
miR-208a MED13 Heart/systemic Energy expenditure 75
miR-133 Prdm16 Skeletal muscle progenitor cells Conversion to brown adipocytes, 78
whole-body thermogenesis
Vessel wall inflammation/
atherosclerosis
miR-126 VCAM-1, RGS16, SPRED1, ECs, arteries? Leukocyte adherence, 52, 53
PIK3R2/p85 atherosclerotic lesion
formation, CXCL12 production
miR-217 SIRTI, FOXO1 ECs Senescence 86
miR-155, miR-342-5p Bcl6, NFB Monocytes, macrophages, CCL2 expression, inflammatory 54 59
neutrophils, ECs, SMCs cell fate, angiotensin II receptor
expression
miR-143/145 KLF2, KLF4, Elk-1, ACE, ACE ECs, SMCs SMC proliferation and 54, 60 63
receptor differentiation
miR-223 P2Y12 Platelets Platelet activation and 64
aggregation
miR-200b PRKAR2B Platelets PKA activity, platelet 65
responsiveness
Cardiac injury/remodeling
after MI
miR-21 Spry1, PTEN Fibroblasts, SMCs Interstitial fibrosis, cardiac 69 71
hypertrophy and dysfunction,
cardiac remodeling, SMC
proliferation and apoptosis,
neointima formation
miR-208a Thyroid hormone-associated Cardiomyocytes Cardiac hypertrophy and 7274
protein 1, myostatin, MYH7, remodeling
miR-499
miR-133/133a1/2 RhoA, Cdc42 Cardiomyocytes Cardiac hypertrophy, cardiac 76
malformation, cardiac fibrosis,
heart failure
miR-92a Integrin-5, eNOS, KLF2, KLF4 ECs Angiogenesis, neovascularization, 80, 81
ventricular function,
cardiomyocyte apoptosis,
infarct size, atherogenesis
miR-126 SPRED1, PI3KS ECs Vascular integrity, angiogenesis 82, 83
miR-221/222 ECs Cell migration, angiogenesis 87
miR-24 GATA2, p21-activated kinase, ECs Apoptosis, vascularity, infarct size, 88
PAK4, BAD/Bcl-XL/Bcl-2 angiogenesis after MI
miR-320 Hsp20 Cardiomyocytes Apoptosis, infarct size post- 89
ischemic injury
miR-199b Dyrk1a ? Calcineurin/NFAT signaling, 90
cardiac hypertrophy, fibrosis,
heart failure
miR-15 Cell cycle checkpoint kinase 1 Cardiomyocytes Cardiomyocyte proliferation, post- 58, 91
ischemic injury or MI

Abbreviations: NPC1, Niemann-Pick disease type C1; Angptl3, angiopoietin-like 3; Gpam, glycerol-3-phosphate acyltransferase 1; MED13, mediator
complex subunit 13; Prdm16, PR domain containing 16; PKA, protein kinase A; SPRED1 Sprouty-related, EVH1 domain-containing protein 1; PIK3R2,
phosphoinositide-3-kinase, regulatory subunit 2; SIRTI, sirtuin 1; FOXO1, Forkhead box protein O1; Spry1, sprouty homolog 1; RhoA, Ras homolog gene
family, member A; Cdc42, cell division control protein 42 homolog; GATA2, GATA binding protein 2; BAD, Bcl-2-associated death promoter; Bcl, B-cell
leukemia/lymphoma; Hsp20, heat shock protein 20; Dyrk1a, dual specificity tyrosine-phosphorylation-regulated kinase 1A.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
4008 Karunakaran and Rayner miRNA and Cardiovascular Health Endocrinology, November 2013, 154(11):4000 4009

References non-human primates raises plasma HDL and lowers VLDL triglyc-
erides. Nature. 2011;478(7369):404 407.
1. Tobert JA. Lovastatin and beyond: the history of the HMG-CoA 26. de Aguiar Vallim TQ, Tarling EJ, Kim T, et al. MicroRNA-144
reductase inhibitors. Nat Rev Drug Discov. 2003;2(7):517526. regulates hepatic ATP binding cassette transporter A1 and plasma
2. Bornfeldt KE, Tabas I. Insulin resistance, hyperglycemia, and ath- high-density lipoprotein after activation of the nuclear receptor
erosclerosis. Cell Metab. 2011;14(5):575585. farnesoid X receptor. Circ Res. 2013;112(12):16021612.
3. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic 27. Ramrez CM, Rotllan N, Vlassov AV, et al. Control of cholesterol
gene lin-4 encodes small RNAs with antisense complementarity to metabolism and plasma high-density lipoprotein levels by mi-
lin-14. Cell. 1993;75(5):843 854. croRNA-144. Circ Res. 2013;112(12):15921601.
4. Pasquinelli AE, Reinhart BJ, Slack F, et al. Conservation of the se- 28. Esau C, Davis S, Murray SF, et al. miR-122 regulation of lipid me-
quence and temporal expression of let-7 heterochronic regulatory tabolism revealed by in vivo antisense targeting. Cell Metab. 2006;
RNA. Nature. 2000;408(6808):86 89. 3(2):8798.
5. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identifica- 29. Lanford RE, Hildebrandt-Eriksen ES, Petri A, et al. Therapeutic
tion of novel genes coding for small expressed RNAs. Science. 2001; silencing of microRNA-122 in primates with chronic hepatitis C
294(5543):853 858. virus infection. Science. 2010;327(5962):198 201.
6. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny 30. Krtzfeldt J, Rajewsky N, Braich R, et al. Silencing of microRNAs
RNAs with probable regulatory roles in Caenorhabditis elegans. in vivo with antagomirs. Nature. 2005;438(7068):685 689.
Science. 2001;294(5543):858 862. 31. Vickers KC, Shoucri BM, Levin MG, et al. MicroRNA-27b is a
7. Lee RC, Ambros V. An extensive class of small RNAs in Caeno- regulatory hub in lipid metabolism and is altered in dyslipidemia.
rhabditis elegans. Science. 2001;294(5543):862 864. Hepatology. 2013;57(2):533542.
8. Claverie JM. Fewer genes, more noncoding RNA. Science. 2005; 32. Mendelsohn ME, Karas RH. The protective effects of estrogen on the
309(5740):1529 1530. cardiovascular system. N Engl J Med. 1999;340(23):18011811.
9. Pasquinelli AE. MicroRNAs and their targets: recognition, regula- 33. Knowlton AA, Lee AR. Estrogen and the cardiovascular system.
tion and an emerging reciprocal relationship. Nat Rev Genet. 2012; Pharmacol Ther. 2012;135(1):54 70.
13(4):271282. 34. Knowlton AA. Estrogen and cardiovascular disease: aging and estrogen
10. Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A. Identification loss at the heart of the matter? Future Cardiol. 2012;8(1):9 12.
of mammalian microRNA host genes and transcription units. Ge- 35. Darblade B, Pendaries C, Krust A, et al. Estradiol alters nitric oxide
nome Res. 2004;14(10A):19021910. production in the mouse aorta through the -, but not -, estrogen
11. Mah SM, Buske C, Humphries RK, Kuchenbauer F. miRNA*: a receptor. Circ Res. 2002;90(4):413 419.
passenger stranded in RNA-induced silencing complex? Crit Rev 36. Nakamura Y, Suzuki T, Miki Y, et al. Estrogen receptors in ath-
Eukaryot Gene Expr. 2010;20(2):141148. erosclerotic human aorta: inhibition of human vascular smooth
12. Giraldez AJ, Mishima Y, Rihel J, et al. Zebrafish MiR-430 promotes muscle cell proliferation by estrogens. Mol Cell Endocrinol. 2004;
deadenylation and clearance of maternal mRNAs. Science. 2006; 219(12):1726.
312(5770):7579. 37. Chan GH, Fiscus RR. Exaggerated production of nitric oxide (NO)
13. Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of and increases in inducible NO-synthase mRNA levels induced by the
mRNA. Proc Natl Acad Sci U S A. 2006;103(11):4034 4039. pro-inflammatory cytokine interleukin-1 in vascular smooth mus-
14. Lee S, Vasudevan S. Post-transcriptional stimulation of gene expres- cle cells of elderly rats. Exp Gerontol. 2004;39(3):387394.
sion by microRNAs. Adv Exp Med Biol. 2013;768:97126. 38. Wilson PW, Garrison RJ, Castelli WP. Postmenopausal estrogen use,
15. Graff JW, Dickson AM, Clay G, McCaffrey AP, Wilson ME. Iden- cigarette smoking, and cardiovascular morbidity in women over 50.
tifying functional microRNAs in macrophages with polarized phe- The Framingham Study. N Engl J Med. 1985;313(17):1038 1043.
notypes. J Biol Chem. 2012;287(26):21816 21825. 39. Yamagata K, Fujiyama S, Ito S, et al. Maturation of microRNA is
16. Heinecke JW. Oxidants and antioxidants in the pathogenesis of hormonally regulated by a nuclear receptor. Mol Cell. 2009;36(2):
atherosclerosis: implications for the oxidized low density lipopro- 340 347.
tein hypothesis. Atherosclerosis. 1998;141(1):115. 40. Zhao J, Imbrie GA, Baur WE, et al. Estrogen receptor-mediated
17. Moore KJ, Tabas I. Macrophages in the pathogenesis of atheroscle- regulation of microRNA inhibits proliferation of vascular smooth
rosis. Cell. 2011;145(3):341355. muscle cells. Arterioscler Thromb Vasc Biol. 2013;33(2):257265.
18. Rader DJ, Tall AR. The not-so-simple HDL story: Is it time to revise 41. Ashcroft FM, Rorsman P. Diabetes mellitus and the -cell: the last
the HDL cholesterol hypothesis? Nat Med. 2012;18(9):1344 1346. ten years. Cell. 2012;148(6):1160 1171.
19. Rayner KJ, Surez Y, Dvalos A, et al. MiR-33 contributes to the 42. El Ouaamari A, Baroukh N, Martens GA, Lebrun P, Pipeleers D, van
regulation of cholesterol homeostasis. Science. 2010;328(5985): Obberghen E. miR-375 targets 3-phosphoinositide-dependent pro-
1570 1573. tein kinase-1 and regulates glucose-induced biological responses in
20. Najafi-Shoushtari SH, Kristo F, Li Y, et al. MicroRNA-33 and the pancreatic -cells. Diabetes. 2008;57(10):2708 2717.
SREBP host genes cooperate to control cholesterol homeostasis. Sci- 43. Poy MN, Hausser J, Trajkovski M, et al. miR-375 maintains normal
ence. 2010;328(5985):1566 1569. pancreatic - and -cell mass. Proc Natl Acad Sci U S A. 2009;
21. Gerin I, Clerbaux LA, Haumont O, et al. Expression of miR-33 from 106(14):58135818.
an SREBP2 intron inhibits cholesterol export and fatty acid oxida- 44. ZhaoH,GuanJ,LeeHM,etal.Up-regulatedpancreatictissuemicroRNA-
tion. J Biol Chem. 2010;285(44):3365233661. 375 associates with human type 2 diabetes through -cell deficit and islet
22. Rayner KJ, Sheedy FJ, Esau CC, et al. Antagonism of miR-33 in mice amyloid deposition. Pancreas. 2010;39(6):843846.
promotes reverse cholesterol transport and regression of atheroscle- 45. Tattikota SG, Sury MD, Rathjen T, et al. Argonaute2 regulates the
rosis. J Clin Invest. 2011;121(7):29212931. pancreatic -cell secretome. Mol Cell Proteomics. 2013;12(5):
23. Horie T, Baba O, Kuwabara Y, et al. MicroRNA-33 deficiency 1214 1225.
reduces the progression of atherosclerotic plaque in ApoE/ mice. 46. Brunham LR, Kruit JK, Pape TD, et al. -Cell ABCA1 influences
J Am Heart Assoc. 2012;1(6):e003376. insulin secretion, glucose homeostasis and response to thiazolidin-
24. Horie T, Ono K, Horiguchi M, et al. MicroRNA-33 encoded by an edione treatment. Nat Med. 2007;13(3):340 347.
intron of sterol regulatory element-binding protein 2 (Srebp2) regulates 47. Wijesekara N, Zhang LH, Kang MH, et al. miR-33a modulates
HDL in vivo. Proc Natl Acad Sci U S A. 2010;107(40):1732117326. ABCA1 expression, cholesterol accumulation, and insulin secretion
25. Rayner KJ, Esau CC, Hussain FN, et al. Inhibition of miR-33a/b in in pancreatic islets. Diabetes. 2012;61(3):653 658.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.
doi: 10.1210/en.2013-1299 endo.endojournals.org 4009

48. Dvalos A, Goedeke L, Smibert P,et al. miR-33a/b contribute to the remodeling occurs in the absence of microRNA-21 in mice. J Clin
regulation of fatty acid metabolism and insulin signaling. Proc Natl Invest. 2010;120(11):39123916.
Acad Sci U S A. 2011;108(22):92329237. 71. Ji R, Cheng Y, Yue J, et al. MicroRNA expression signature and
49. Trajkovski M, Hausser J, Soutschek J, et al. MicroRNAs 103 and antisense-mediated depletion reveal an essential role of MicroRNA
107 regulate insulin sensitivity. Nature. 2011;474(7353):649 653. in vascular neointimal lesion formation. Circ Res. 2007;100(11):
50. Herrera BM, Lockstone HE, Taylor JM, et al. Global microRNA 1579 1588.
expression profiles in insulin target tissues in a spontaneous rat 72. Callis TE, Pandya K, Seok HY, et al. MicroRNA-208a is a regulator
model of type 2 diabetes. Diabetologia. 2010;53(6):1099 1109. of cardiac hypertrophy and conduction in mice. J Clin Invest. 2009;
51. Jordan SD, Krger M, Willmes DM, et al. Obesity-induced overex- 119(9):27722786.
pression of miRNA-143 inhibits insulin-stimulated AKT activation 73. van Rooij E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN.
and impairs glucose metabolism. Nat Cell Biol. 2011;13(4):434 446. Control of stress-dependent cardiac growth and gene expression by
52. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowenstein CJ. a microRNA. Science. 2007;316(5824):575579.
MicroRNA-126 regulates endothelial expression of vascular cell ad- 74. Montgomery RL, Hullinger TG, Semus HM, et al. Therapeutic in-
hesion molecule 1. Proc Natl Acad Sci U S A. 2008;105(5):1516 1521. hibition of miR-208a improves cardiac function and survival during
53. Zernecke A, Bidzhekov K, Noels H, et al. Delivery of microRNA- heart failure. Circulation. 2011;124(14):15371547.
126 by apoptotic bodies induces CXCL12-dependent vascular pro- 75. Grueter CE, van Rooij E, Johnson BA, Det al. A cardiac microRNA
tection. Sci Signal. 2009;2(100):ra81. governs systemic energy homeostasis by regulation of MED13. Cell.
54. Fichtlscherer S, De Rosa S, Fox H, et al. Circulating microRNAs in 2012;149(3):671 683.
patients with coronary artery disease. Circ Res. 2010;107(5):677 76. Car A, Catalucci D, Felicetti F, et al. MicroRNA-133 controls car-
684. diac hypertrophy. Nat Med. 2007;13(5):613 618.
55. Nazari-Jahantigh M, Wei Y, Noels H, et al. MicroRNA-155 pro- 77. Liu N, Bezprozvannaya S, Williams AH, et al. microRNA-133a regu-
motes atherosclerosis by repressing Bcl6 in macrophages. J Clin lates cardiomyocyte proliferation and suppresses smooth muscle gene
Invest. 2012;122(11):4190 4202. expression in the heart. Genes Dev. 2008;22(23):32423254.
56. Donners MM, Wolfs IM, Stoger LJ, et al. Hematopoietic miR155 78. Matkovich SJ, Wang W, Tu Y, et al. MicroRNA-133a protects
deficiency enhances atherosclerosis and decreases plaque stability in against myocardial fibrosis and modulates electrical repolarization
hyperlipidemic mice. PLoS One. 2012;7(4):e35877. without affecting hypertrophy in pressure-overloaded adult hearts.
Circ Res. 2010;106(1):166 175.
57. Wei Y, Nazari-Jahantigh M, Chan L, et al. The microRNA-3425p
79. Yin H, Pasut A, Soleimani VD, et al. MicroRNA-133 controls
fosters inflammatory macrophage activation through an Akt1- and
brown adipose determination in skeletal muscle satellite cells by
microRNA-155-dependent pathway during atherosclerosis. Circu-
targeting Prdm16. Cell Metab. 2013;17(2):210 224.
lation. 2013;127(15):1609 1619.
80. Bonauer A, Carmona G, Iwasaki M, et al. MicroRNA-92a controls
58. Porrello ER, Johnson BA, Aurora AB, et al. MiR-15 family regulates
angiogenesis and functional recovery of ischemic tissues in mice.
postnatal mitotic arrest of cardiomyocytes. Circ Res. 2011;109(6):
Science. 2009;324(5935):1710 1713.
670 679.
81. Fang Y, Davies PF. Site-specific microRNA-92a regulation of Krup-
59. Zhu N, Zhang D, Chen S, et al. Endothelial enriched microRNAs
pel-like factors 4 and 2 in atherosusceptible endothelium. Arterio-
regulate angiotensin II-induced endothelial inflammation and mi-
scler Thromb Vasc Biol. 2012;32(4):979 987.
gration. Atherosclerosis. 2011;215(2):286 293.
82. Wang S, Aurora AB, Johnson BA, et al. The endothelial-specific
60. Cordes KR, Sheehy NT, White MP, et al. miR-145 and miR-143
microRNA miR-126 governs vascular integrity and angiogenesis.
regulate smooth muscle cell fate and plasticity. Nature. 2009;
Dev Cell. 2008;15(2):261271.
460(7256):705710. 83. Fish JE, Santoro MM, Morton SU, et al. miR-126 regulates angio-
61. Boettger T, Beetz N, Kostin S, et al. Acquisition of the contractile genic signaling and vascular integrity. Dev Cell. 2008;15(2):272
phenotype by murine arterial smooth muscle cells depends on the 284.
Mir143/145 gene cluster. J Clin Invest. 2009;119(9):2634 2647. 84. Broderick JA, Zamore PD. MicroRNA therapeutics. Gene Ther.
62. Lovren F, Pan Y, Quan A, et al. MicroRNA-145 targeted therapy 2011;18(12):1104 1110.
reduces atherosclerosis. Circulation. 2012;126(11 Suppl 1):S81 85. Elmn J, Lindow M, Schtz S, et al. LNA-mediated microRNA silenc-
S90. ing in non-human primates. Nature. 2008;452(7189):896 899.
63. Hergenreider E, Heydt S, Trguer K, et al. Atheroprotective com- 86. Franciscus A. Santaris Pharma A/S advances miravirsen, the first
munication between endothelial cells and smooth muscle cells microRNA-targeted drug to enter clinical trials, into Phase 2 to treat
through miRNAs. Nat Cell Biol. 2012;14(3):249 256. patients infected with Hepatitis C virus. HCV Advocate website.
64. Landry P, Plante I, Ouellet DL, Perron MP, Rousseau G, Provost P. http://www.hcvadvocate.org/hepatitis/hepc/HCVDrugs.html. Ac-
Existence of a microRNA pathway in anucleate platelets. Nat Struct cessed March 21, 2011.
Mol Biol. 2009;16(9):961966. 87. Poliseno L, Tuccoli A, Mariani L, et al. MicroRNAs modulate the
65. Nagalla S, Shaw C, Kong X, et al. Platelet microRNA-mRNA co- angiogenic properties of HUVECs. Blood. 2006;108(9):3068
expression profiles correlate with platelet reactivity. Blood. 2011; 3071.
117(19):5189 5197. 88. Fiedler J, Jazbutyte V, Kirchmaier BC, et al. MicroRNA-24 regulates
66. Diehl P, Fricke A, Sander L, et al. Microparticles: major transport vascularity after myocardial infarction. Circulation. 2011;124(6):
vehicles for distinct microRNAs in circulation. Cardiovasc Res. 720 730.
2012;93(4):633 644. 89. Ren XP, Wu J, Wang X, et al. MicroRNA-320 is involved in the
67. Willeit P, Zampetaki A, Dudek K, et al. Circulating microRNAs as regulation of cardiac ischemia/reperfusion injury by targeting heat-
novel biomarkers for platelet activation. Circ Res. 2013;112(4): shock protein 20. Circulation. 2009;119(17):23572366.
595 600. 90. da Costa Martins PA, Salic K, Gladka MM, et al. MicroRNA-199b
68. Jazbutyte V, Thum T. MicroRNA-21: from cancer to cardiovascu- targets the nuclear kinase Dyrk1a in an auto-amplification loop
lar disease. Curr Drug Targets. 2010;11(8):926 935. promoting calcineurin/NFAT signalling. Nat Cell Biol. 2010;
69. Thum T, Gross C, Fiedler J, et al. MicroRNA-21 contributes to 12(12):1220 1227.
myocardial disease by stimulating MAP kinase signalling in fibro- 91. Hullinger TG, Montgomery RL, Seto AG, et al. Inhibition of miR-15
blasts. Nature. 2008;456(7224):980 984. protects against cardiac ischemic injury. Circ Res. 2012;110(1):
70. Patrick DM, Montgomery RL, Qi X, et al. Stress-dependent cardiac 71 81.

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 05 July 2016. at 23:43 For personal use only. No other uses without permission. . All rights reserved.

You might also like