You are on page 1of 22

SCIENCE TRANSLATIONAL MEDICINE | REVIEW

MYOCARDIAL INFARCTION Copyright © 2021


The Authors, some
Cell type–specific microRNA therapies for rights reserved;
exclusive licensee
myocardial infarction American Association
for the Advancement
of Science. No claim
Bohao Liu1,2*, Bryan Wang1,2*, Xiaokan Zhang1, Roberta Lock2, to original U.S.
Trevor Nash1,2, Gordana Vunjak-Novakovic1,2† Government Works

Current interventions fail to recover injured myocardium after infarction and prompt the need for development
of cardioprotective strategies. Of increasing interest is the therapeutic use of microRNAs to control gene expression
through specific targeting of mRNAs. In this Review, we discuss current microRNA-based therapeutic strategies,
describing the outcomes and limitations of key microRNAs with a focus on target cell types and molecular pathways.
Last, we offer a perspective on the outlook of microRNA therapies for myocardial infarction, highlighting the out-
standing challenges and emerging strategies.

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


INTRODUCTION miRNAs are incorporated into miRNA-induced silencing complexes,
Heart disease remains as the leading cause of mortality worldwide, the functional unit that targets mRNAs with near-perfect base pair-
with myocardial infarction (MI) affecting more than 700,000 indi- ing to inhibit gene expression (5). Known to interact with the
viduals annually in the United States alone (1). Because the adult majority of mammalian protein-coding genes, miRNAs are powerful
heart lacks ability to innately repair and regenerate after injury, MI mediators of a diverse spectrum of processes, both during normal
results in permanent loss of myocardial cells (2). The damaged heart heart development and physiology and during cardiovascular dis-
undergoes pathological remodeling, leading to reduced contractile ease progression (6). miRNAs are implicated in every phase of MI
function and often heart failure (3). Standard therapies mainly focus progression; in response to ischemia, miRNAs in both mouse and
on revascularizating the occluded artery to salvage as much of the human hearts have been shown to be dysregulated, contributing to
myocardium as possible but fail to adequately recover injured myo- the progression of many pathological processes.
cardium. Any advances in the treatment of MI would thus have major For treatment of MI, miRNAs represent particularly attractive
clinical significance. therapeutic targets due to several unique characteristics. Most im-
The human heart is composed of an array of different cell types portant is the pleotropic ability of a single miRNA to regulate multi-
including cardiomyocytes (CMs), cardiac fibroblasts (CFs), endo- ple pathologically disrupted biological pathways across different cell
thelial cells (ECs), and immune cells. Each cell type contributes to types (7). This is in stark contrast to traditional drug-based ap-
cardiac function in a distinct way, with intercellular communication proaches that target singular molecules and pathways. Such a pleio-
and coordination being vital to maintaining normal organ function tropic approach is especially powerful for treatment of MI because
(Fig. 1). The pathological changes in cardiac remodeling after MI injury is not instigated by a single genetic link or biological process
involve each of these cell types and multiple molecular mechanisms but rather by multiple coordinated processes. In addition, miRNAs
during four partially overlapping phases: the inflammatory, prolif- are ideal therapeutic targets because they are small, precisely defined
erative, maturation, and remodeling phases (3). The inflammatory nucleic sequences for which mimics or antisense oligonucleotides
phase is initiated by massive cell death in the infarct area. Although (ASOs) can effectively and efficiently be designed with high affinity
CFs degrade the extracellular matrix (ECM), changes in EC-mediated and specificity. Last, when compared to cell-based approaches,
vascular permeability allow immune cells to migrate into the injured miRNA therapies may provide similar benefits without the challenges
area and facilitate clearance of damaged cells. In the proliferative of immune rejection and poor cell engraftment.
phase, inflammation is dampened by macrophage phenotypic switch- Although miRNA therapies have potential for treating MI, a major
ing, and reparative processes begin. Fibroblasts and ECs proliferate, challenge that limits their clinical advancement is a lack of under-
deposit collagen, and establish a new microvascular network in the standing of the molecular mechanisms behind their therapeutic
infarct area. In the maturation phase, ECM proteins secreted by acti- properties. Specifically, the pleotropic effects of a miRNA on each
vated fibroblasts undergo cross-linking to form a stable scar. Last, cell type and on different biological pathways within the heart must
during long-term remodeling, maladaptive processes including be carefully delineated to facilitate clinical translation. In this Review,
cardiomyocyte pathological hypertrophy, remote fibrosis, and cap- we examine recently published studies describing the therapeutic
illary rarefaction occur to contend with the stresses arising from the manipulation of miRNAs in the treatment of MI. We describe the
failing heart (Fig. 1). effect of miRNAs on each cell type and how miRNAs directly target
Recently, microRNAs (miRNAs) have emerged as a promising molecular pathways to modulate cell type–specific behavior. Last, we
therapy for MI. miRNAs are small noncoding RNAs that regulate gene discuss key prospects and challenges for developing miRNA therapies.
expression at the posttranscriptional level (4). Mature, single-stranded

1
CHARACTERISTICS OF miRNA THERAPIES
Department of Medicine, Columbia University, New York, NY 10032, USA. 2Depart- Focusing on literature within the past 3 years and foundational
ment of Biomedical Engineering, Columbia University, New York, NY 10032, USA.
*These authors contributed equally to this work. studies from the past 10 years, we identified a total of 213 relevant
†Corresponding author. Email: gv2131@columbia.edu studies detailing 116 unique miRNAs (Fig. 2). Therapeutic miRNA

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 1 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

Remodeling Infarct zone Cardiomyocytes Fibroblasts Endothelial cells Immune cells


phase responses

Healthy heart Provide the Maintain Serve as a barrier Mediate


contractile extracellular between blood and inflammatory
apparatus for the matrix (ECM); myocardial tissue; response to heart
Healthy cardiomyocytes myocardium; have sense damage; regulate vascular injury with
Healthy vessel high metabolic quiescent. tone and preformed stores
Resident immune cell demands; permeability. of inflammatory
nonproliferate in mediators.
Cardiac fibroblast the adult heart.

Inflammatory phase Irreversibly injured Undergo activation Undergo activation Infiltrate the
by ischemia, leading leading to secretion leading to increased infarcted
to cell death through of cytokines and permaeability, area; express
Dying cardiomyocytes multiple pathways MMPs; responsible surface adhesion proinflammatory
Infarcted coronary including necrosis, for ECM degradation. molecules chemokines/
apoptosis, and expression, cytokines;

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


autophagy; initiate recruitment clear cellular debris.
Activated proinflammatory inflammation of immune cells.
Myocardial infarction progression

fibroblast through cytokine


Infiltrated M1 macrophage release.

Proliferative phase Undergo very Myofibroblast and Undergo Macrophages


limited, if any, smooth muscle angiogenesis, undergo
Activated myofibroblasts proliferation. gene activation; which contributes to polarization,
proliferation into revascularization switching toward
Collagen fibers infarct zone; ECM and remodeling. reparative
Endothelial cell synthesis. phenotype; secrete
anti-inflammatory
signals for repair
response.
Polarized M2 macrophage

Maturation phase Often hypertrophy Mediate ECM Undergo Undergo apoptotic


with the addition of cross-linking; scar angiogenesis cell death in the
Collagen-based scar new myofibrils. contraction; and proliferation to infarct zone.
transition form vessel lumens
to matrifibrocyte and eventually are
Newly formed vessel phenotype. stabilized by
recruited pericytes.
Cardiomyocytes hypertrophy

Long-term remodeling Undergo further Chronic Microvascular Reemergence of


pathologic overactivation, rarefaction and immune cells leads
Remote fibrosis hypertrophy to leading to fibrosis reduction of vascular to inflammation in
adapt to the physical and increased tissue density in the heart. remote,
demands on the stiffness in both noninfarcted
Microvascular rarefaction injured heart. infarcted and remote myocardium.
myocardium.
Cardiomyocytes hypertrophy

Remote inflammation

Fig. 1. Summary of myocardial infarct progression with a focus on the role of each cell type. Immediately after infarction, the inflammatory phase begins and is
characterized by irreversible cell death of all cell types in the infarct and the initiation of the inflammatory process through cytokine release. Next, the proliferative phase
is characterized by the activation of CFs and the initiation of scar development. Macrophages also undergo polarization and switch to an anti-inflammatory phenotype
during this phase. The maturation phase is characterized by cardiomyocyte hypertrophy in response to increased cardiac demand and the maturation of the scar that
forms over the infarcted area. Last, long-term remodeling is characterized by pathological changes including maladaptive cardiomyocyte hypertrophy, chronic overacti-
vation of CFs, decreased vascular density, and reemergence of an inflammatory response.

targets were identified by two methods: (i) miRNA profiling after Using these characteristics, we assigned a clinical applicability
MI, which revealed substantial dysregulation of miRNAs natively score representing a qualitative measure of the evidence supporting
CREDIT: A. KITTERMAN/SCIENCE TRANSLATIONAL MEDICINE

expressed in the heart; and (ii) miRNA screening in in vitro injury the therapeutic application of each miRNA reviewed. This score is
models, which identified potential proregenerative miRNAs not denoted by the footnotes in Table 1. “*” indicates therapeutic efficacy
natively expressed in the heart. To understand cell type–specific ef- in in vitro cell-based models; “**” indicates therapeutic efficacy in
fects of miRNA therapies, we organized the reviewed miRNAs by the in vivo models with target pathway identification and characteriza-
cell types they target. For each study, we extracted a set of character- tion; and “***” indicates therapeutic efficacy in multiple model systems
istics describing each miRNA. miRNAs were defined as beneficial if and well-defined mechanisms of action including comprehensively
treatment with miRNA mimics resulted in improved recovery after validated direct targets. Table 1 presents a select list of key miRNAs for
MI and as harmful if treatment with miRNA inhibitors resulted in each cell type. The complete list of surveyed miRNAs is presented in
improved recovery after MI. We noted the model systems each table S1 and summarized in table S2.
study used in vitro and in vivo as well as the delivery of the miRNA Most miRNAs play both beneficial and harmful roles in the pro-
therapies (miRNA formulation and the method of administration). gression of MI. In addition, a majority of miRNAs affect multiple
For each miRNA studied, we detail the target pathway and the spe- cell types, thus demonstrating the important pleiotropic effects of
cific target molecule. miRNAs when used in cardiac therapy. Here, we first highlight

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 2 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

miRNA effect miRNA pleiotropy Target cell type Target process receptors on the cell surface such as Fas ligand activating Fas receptor
(FasR) and tumor necrosis factor– (TNF) activating TNF receptor 1
(TNFR1) (9).
Beneficial: 79
Single target cell
Autophagy
Apoptosis: 101
type: 109 In response to the ischemic or metabolic stress during MI, cardio-
Cardiomyocyte: 132
myocytes can activate the process of autophagy, which allows them
Unique miRNAs: 116

to catabolize damaged or dysfunctional macromolecular structures


Studies: 213

Harmful: 43 by lysosomal degradation to maintain cellular homeostasis. Primary


Proliferation: 22 players in cardiomyocyte autophagy include the mammalian target
Autophagy: 11 of rapamycin (mTOR) and AMP-activated protein kinase (AMPK)
Inflammatory cell: 17 Hypertrophy: 4
Arrhythmia: 2 pathways (10). Although undoubtedly critical in cardiomyocyte re-
Multiple target cell
Inflammation: 21 sponse to MI, the role of autophagy in mediating the extent of myo-
Endothelial cell: 24
Both: 91 types: 104 Necrosis: 6 cardial injury is uncertain.
Angiogenesis: 19 Proliferation and remodeling

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Fibroblast: 26
Historically, adult cardiomyocytes have been described as post-
Fibrosis: 22
Not specified: 14 mitotic, but this paradigm has shifted, with studies indicating that
Other/not specified: 5
cardiomyocytes proliferate to a small extent in the adult myocardium
Fig. 2. Summary of miRNAs surveyed in this Review. For each study, in this (11). Although limited, exogenously promoted cardiomyocyte pro-
Review, we categorized the overall effect of the miRNA studied, whether the miRNA liferation has become a potential therapeutic strategy for heart failure.
was shown to affect multiple cell types (pleiotropy), which cell types were studied,
Over the past decade, several promising targets have emerged that
and the physiologic process that the miRNA was shown to target. The numbers
following each division represents the number of studies that is in that category.
induce cardiomyocyte proliferation, including Hippo–Yes-associated
protein (YAP), homeodomain-only protein (HOPX), and follistatin-­
related protein 1 (FSTL) (12–14).
post-MI events by cell type, briefly describing how key processes in Because of the loss of functional cells after MI, cardiomyocytes
each cell type affect their phenotype and function. After providing undergo hypertrophy and morphological changes to adapt to the
this contextual framework, we examine the recent advances in miRNA physical demands of the injured heart. Although initially compen-
therapies targeting each cell type. satory, prolonged increases in functional demands eventually lead to
pathological remodeling and a decrease in cardiomyocyte function. In-
farcted tissue also interrupts gap junctions between cardiomyocytes,
ROLE OF miRNAs IN CARDIOMYOCYTE RESPONSE causing aberrant electrical conduction leading to arrhythmias (15).
TO MYOCARDIAL INFARCTION
Cardiomyocytes are the functional cells of the heart, responsible for Recent advances in miRNA therapies targeting
generating contractile force. Because of their large energy demands, cardiomyocytes
cardiomyocytes are highly susceptible to death upon loss of blood Most studies (132 of 213 studies) describing the therapeutic appli-
supply. Cardiomyocytes have limited proliferative ability, and those cation of miRNAs to MI have cardiomyocytes as the primary target
lost during MI cannot be replaced (8). Thus, two major approaches cell type. These studies have focused on identifying and using miRNAs
to therapeutically target cardiomyocytes after MI are prevention of capable of either preventing cardiomyocyte death (88 of 132 studies)
cardiomyocyte cell death after ischemic stress and induction of or promoting cardiomyocyte proliferation (18 of 132 studies). Here,
cardiomyocyte proliferation after resolution of injury. we summarize key therapeutic miRNAs and their mechanisms of action
in cardiomyocytes (Fig. 3).
Cardiomyocyte response to myocardial infarction Apoptosis
Apoptosis and necrosis Many miRNAs have been identified that regulate components of
The primary pathways of regulated cardiomyocyte cell death during apoptosis pathways, with beneficial miRNAs inhibiting these pathways
MI are apoptosis and necrosis, both of which have distinct molecular to prevent cardiomyocyte apoptosis and harmful miRNAs inducing
mechanisms and cellular conditions. Mechanistically, apoptosis and apoptosis after injury. In early apoptosis, mitochondrial fission leads
necrosis can both be subdivided into mitochondria- and receptor-­ to activation of mitochondria-mediated cardiomyocyte death. In a series
CREDIT: A. KITTERMAN/SCIENCE TRANSLATIONAL MEDICINE

mediated pathways, known as the intrinsic and extrinsic pathways of studies, Wang and colleagues (16) described five miRNAs that target
during apoptosis and as mitochondrial necrosis and necroptosis components of mitochondrial fission machinery. They identified two
during necrosis. During MI, the intrinsic pathway is initiated when beneficial miRNAs, miR-499 and miR-652, which prevent mitochondrial
free radical generation and low adenosine triphosphate (ATP) lead fission. miR-499 directly inhibits both the  and  isoforms of the
to mitochondrial stress and subsequent perturbation of the B cell calcineurin catalytic subunit, thereby decreasing the accumulation of
lymphoma 2 (BCL-2) family of proteins. Simultaneously, mito- dynamin-related protein 1 (DRP1), a guanosine triphosphatase required
chondrial necrosis is initiated when intracellular Ca2+ overload alters for mitochondrial fission (16). miR-652 inhibits the proapoptotic
the dynamics of the mitochondrial membrane, leading to a disrup- mitochondrial membrane protein MTP18 (17). In contrast, miR-361
tion in permeability. This results in a rapid decrease in ATP, which and miR-539 were identified as harmful miRNAs that inhibit pro-
prevents the cell from carrying out necessary repair functions, lead- hibitins 1 and 2, respectively, increasing mitochondrial fission and
ing to organelle dysfunction and plasma membrane rupture. The apoptosis after MI (18, 19). miR-421 was also identified as harmful
extrinsic pathway of apoptosis and necroptosis are both triggered after MI as it directly targets the mitochondrial Ser/Thr kinase PTEN
during MI by release of stimuli outside the cell that activates death induced kinase 1 (PINK1), which inhibits mitochondrial fission (20).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 3 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

Table 1. Select cell type–specific miRNA therapies in the treatment of MI. Complete list of miRNAs reviewed in table S1. IRI, ischemia-reperfusion injury;
AngII, angiotensin II; TAC, transverse aortic constriction; IV, intravenous; IM, intramyocardial; IP, intraperitoneal; I. Ventr, intraventricular; EV, extracellular vesicle;
Transf, transfection; Lenti, lentiviral; EndMT, endothelial-to-mesenchymal transition; Mito, mitochondrial; ROS, reactive oxygen species.
Delivery Target Target Target Clinical
miRNA Effect Injury model Reference
method process pathway molecule application
Cardiomyocytes
miR-1 Harmful In vivo IRI IV Apoptosis PKC apoptosis PKC ** (43)
In vivo
miR-9 Harmful IM Proliferation FSTL FSTL *** (73)
ischemia
In vivo
miR-22 Harmful IP Autophagy Autophagy PPAR *** (59)
ischemia
In vivo
miR-302/367 Beneficial IV Proliferation Hippo-YAP MST1 *** (69)
ischemia

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


ROS
miR-762 Harmful In vivo IRI IV Apoptosis ND2 *** (49)
production
miR-873 Beneficial In vivo IRI IV Necrosis Necroptosis RIPK1/RIPK3 *** (53)
Let-7a Beneficial In vivo AngII I. Ventr. Hypertrophy Undefined CaM *** (80)
Cardiac fibroblasts
In vivo
miR-21 Harmful IM Fibrosis ERK/MAPK SPRY1 *** (112)
ischemia
miR-21 Harmful In vivo TAC IV Apoptosis ERK/MAPK SPRY1 *** (111)
miR-92a Harmful In vitro TGF- EV Fibrosis TGF- SMAD7 * (106)
In vivo
miR-101 Beneficial I. Ventr. Fibrosis TGF- cFOS *** (96)
ischemia
miR-155 Beneficial In vivo genetic NA Proliferation RAS SOS1 ** (178)
Endothelial cells
miR-32 Harmful In vitro Transf. Proliferation Undefined KLF2 * (228)
In vivo
miR-155 Harmful IM Angiogenesis AMPK RAC1 *** (142)
ischemia
MtFR1
miR-324 Beneficial In vitro H2O2 Transf. Apoptosis Mitofission * (135)
(indirect)
In vivo
miR-532 Beneficial IM Angiogenesis EndMT PRSS23 *** (151)
ischemia
Inflammatory cells
In vivo Inflammatory Intrinsic
miR-24 Beneficial EV BIM *** (161)
ischemia infiltration pathway
In vivo Inflammatory
miR-155 Harmful IV NF-B SOCS1 *** (175)
ischemia activation
Inflammatory PI3k/AKT/
miR-182 Beneficial In vivo IRI EV TLR4 ** (162)
resolution mTOR
In vivo Inflammatory
miR-224 Beneficial Lenti. TGF- Undefined * (157)
ischemia activation

*Indicates therapeutic efficacy in in vitro cell-based models.   **Indicates therapeutic efficacy in in vitro and in vivo models with target pathway identification
and characterization.   ***Indicates therapeutic efficacy in multiple model systems and well-defined mechanisms of action including comprehensively
validated direct targets.

Harmful miRNAs including miR-195 and miR-1 have been shown reducing BIM expression. miRNAs affect other components of the
to directly inhibit the antiapoptotic protein BCL-2 that regulates the intrinsic pathway: For example, miR-125b inhibits proapoptotic
intrinsic pathway (21, 22). Alternatively, beneficial miRNAs such as Bcl-2 homologous antagonist/killer 1 (BAK1), miR-17 inhibits
miR-19b and miR-24 have been shown to directly inhibit the proapop- proapoptotic apoptotic protease activating factor-1 (APAF-1), and
totic protein Bcl-2-like protein 11 (BIM) (23, 24). In a foundational miR-27a inhibits Bcl-2 interaction protein 3 (BNIP3) (25–27). To
study, Qian et al. (24) demonstrated that the overexpression of miR- modulate the extrinsic pathway, miR-133b can down-regulate the
24 via intramyocardial injection of a miR-24 mimic attenuated death receptor FasR, and the effector caspase-3 can be inhibited
ischemia-induced injury and restored cardiac function by directly by miR-1192 (28, 29). Last, miR-327 has been shown to inhibit

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 4 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Fig. 3. Summary of miRNAs that target cardiomyocytes after MI. miRNAs have been shown to regulate apoptosis, necrosis, and autophagy-mediated cardiomyocyte
cell death. For cardiomyocyte proliferation, miRNAs have been shown to modulate multiple proliferative pathways as well as the cell cycle directly. Last, miRNAs have also
been shown to regulate important processes including hypertrophy, arrhythmia, and inflammation. Select well-characterized miRNAs, their targets (written above the
arrows), and the processes that they regulate are shown (i.e., miR-19b prevents apoptosis through BIM signaling). miRNAs prefaced with “anti–” denote that the inhibition
of the miRNA is therapeutic (i.e., the inhibition of miR-1 prevents apoptosis through BCL-2 signaling). For the complete list of miRNAs reviewed that affect cardiomyocytes,
please see table S1.

apoptosis repressor with caspase recruitment domain (ARC), a po- NF-B signaling, miR-145a exerts beneficial effects by inhibiting
tent repressor of the intrinsic and extrinsic signaling pathways (30). early growth response 1 (EGR1)–mediated NF-B activation. In Notch
In cardiomyocytes, multiple pathways respond to ischemic stress signaling, the harmful miRNAs miR-363 and miR-429 directly inhibit
and injury after MI and ultimately determine whether the cell the Notch 1 receptor, preventing Notch-­mediated antiapoptotic sig-
undergoes apoptosis. The PI3k/AKT pathway plays a major role in naling (46, 47). In contrast, miR-322 targets the Notch inhibitor
controlling cell survival and the inhibition of programmed cell death FBXW7, up-regulating Notch signaling and preventing apoptosis
through stimulatory phosphorylation of prosurvival genes and the after MI (48). In the regulation of ROS, miRNAs can directly affect
inhibitory phosphorylation of proapoptotic genes (31). Many miRNAs mitochondrial respiration. For example, the harmful miR-762 inhibits
modulate the PI3k/AKT pathway in cardiomyocytes after MI. Spe- the mitochondrial nicotinamide adenine dinucleotide + hydrogen
cifically, the central AKT inhibitor phosphatase and tensin homo- (NADH) dehydrogenase subunit ND2 leading to increased ROS
log (PTEN) can be directly targeted by miRNAs miR-146b, miR-182, production, whereas the beneficial miR-183 inhibits the mitochon-
miR-26a, miR-494, and miR-93 (32–35). Song et al. (36) demonstrated drial membrane ion channels voltage-dependent anion channel 1
that miR-320 directly inhibits the ligand insulin growth factor 1 (VDAC1) leading to decreased ROS production (49, 50). In addition,
(IGF-1) after ischemia-reperfusion injury (IRI), thus preventing IGF Su et al. (51) showed that miR-132 inhibits histone deacetylase HDAC3,
receptor–mediated activation of the PI3k/AKT pathway. Restoring preventing its ability to up-regulate genes that promote ROS accu-
IGF-1 function using a lentivirus expressing miR-320 inhibitor led mulation in cardiomyocytes.
to a decrease in the number of apoptotic cardiomyocytes and preserved Necrosis
cardiac function (36). Another notable pathway regulating apop- Large numbers of cardiomyocytes undergo necrotic cell death after
CREDIT: A. KITTERMAN/SCIENCE TRANSLATIONAL MEDICINE

tosis centers around the molecule programmed cell death 4 (PDCD4), ischemic injury, providing the main stimulus for postinfarction
which is up-regulated during apoptosis and functions as a proapoptotic inflammatory activation. Here, beneficial miRNAs prevent cardio-
inhibitor of gene transcription and translation. During MI, many myocyte necrosis, whereas harmful miRNAs induce necrosis after
miRNAs directly inhibit PDCD4 and prevent apoptosis in cardio- injury. The central component of necroptosis activation is the as-
myocytes including miR-200, miR-499, miR-532, and, importantly, sembly of the receptor-interacting protein kinase 1 (RIPK1)–RIPK3-­
miR-21, a miRNA with notable pleiotropic effects (37–42). mixed lineage kinase-like (MLKL) signaling complex. Two miRNAs,
Additional pathways implicated in cardiomyocyte apoptosis miR-105 and miR-873, prevent necroptosis through the direct
include protein kinase C (PKC), nuclear factor kappa-light-chain- down-regulation of RIPK proteins (52, 53). In addition, miRNAs
enhancer of activated B cells (NF-B), Notch, and reactive oxygen can modulate the expression of death receptors responsible for ini-
species (ROS) homeostasis. In PKC-mediated apoptosis, the harm- tiating necroptosis. The beneficial miR-223 inhibits the receptors
ful miRNAs miR-1, miR-143, and miR-31 have been shown to TNFR1 and DR6, thereby preventing necroptosis; whereas the
directly target antiapoptotic protein kinase C epsilon (PRKCE), harmful miR-103/107 inhibits the antinecrotic receptor Fas-associated
thereby leading to increased cardiomyocyte apoptosis (43–45). In death domain protein, thereby activating necroptosis (54, 55). Last,

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 5 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

the beneficial miR-874 inhibits caspase-8 preventing its ability to preventing it from activating positive cell cycle regulators cyclin E
activate the RIPK signaling complex (56). and CDK2 (77).
Autophagy Other effects
Studies have identified miRNAs that modulate post-ischemic auto- Beyond cardiomyocyte cell death and proliferation, miRNAs can
phagy, sometimes with contradictory effects on cardiac function. For affect other facets of cardiac physiology. The harmful miRNAs
example, Wang et al. (57) demonstrated that miR-188 suppresses miR-1231 and miR-223 induce arrhythmias in the heart after MI
autophagy-mediated apoptosis after MI through direct inhibition of through inhibition of the ion channels CACNA2D2 and KCND2,
the autophagy-related gene ATG7. Similarly, Liu et al. (58) showed respectively (78, 79). The beneficial miRNAs Let-7a and miR-206 pre-
that miR-93 protects cardiomyocytes from apoptosis through inhi- vent pathological cardiomyocyte hypertrophy by targeting calmodulin
bition of ATG7. In a comprehensive study, Gupta et al. (59) identi- (CAM) and forkhead box protein P1 (FOXP1), respectively (80, 81).
fied miR-22 as a potent inhibitor of cardiac autophagy through its Meanwhile, the harmful miRNAs of the miR-212/132 family can induce
targeting of peroxisome proliferator–activated receptor  (PPAR), hypertrophy through inhibition of FOXO3 and subsequent hyper-
a nuclear receptor known to activate autophagy. However, inhibi- activation of NFAT signaling (60). A series of miRNAs have been shown
tion of miR-22 leading to the activation of autophagy after MI to modulate the ability of cardiomyocytes to activate the inflammatory

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


resulted in improved postinfarction remodeling and improved car- cascade: miR-128 targets SOX7, resulting in increased inflammation;
diac function (59). Similarly, Ucar et al. (60) demonstrated that the miR-135b targets caspase-1, resulting in reduced inflammation; and
miR-212/132 family inhibited autophagy and that their inhibition miR-145 targets CD40, also resulting in reduced inflammation (82–84).
restored the autophagic response, resulting in the rescue of heart
failure in mice. Other studies have identified miRNAs that can
either promote or inhibit autophagy including miR-142, miR-145, ROLE OF miRNAs IN FIBROBLAST RESPONSE
miR-221, miR-301, miR-18a, miR-199a, miR-30a, and miR-558; TO MYOCARDIAL INFARCTION
however, the ultimate therapeutic effects of these miRNAs seem to After MI, CFs become activated and differentiated into myofibro-
depend on their individual targets and not the overall activation or blasts, which play an integral role in the rapid formation of a scar
inactivation of autophagy (61–68). necessary to prevent ventricular wall rupture. However, excessive
Proliferation CF activation, proliferation, and ECM deposition after MI contribute
Cardiac function can be restored after MI by generation of new cardio- to pathological cardiac fibrosis, which can exacerbate injury and lead
myocytes. To this end, multiple miRNAs have been identified that to heart failure. Reversion of this activated phenotype with miRNAs
can regulate cardiomyocyte proliferation. Here, beneficial miRNAs is a key approach toward attenuating this pathological response.
promote, whereas harmful miRNAs prevent cardiomyocyte prolif-
eration after injury. Fibroblast responses to myocardial infarction
The Hippo-YAP pathway is a critical regulator of organ size and Inflammation
growth that plays an important role in cardiomyocyte proliferation Immediately after MI, fibroblasts produce proinflammatory cyto-
(12). In a foundational study that delivered a transient proliferative kines in response to cardiomyocyte death and inflammatory milieu.
stimulus using miRNA mimics, Tian et al. (69) showed that the de- Interleukin-1 (IL-1) stimulates CFs to secrete TNF and IL-1
livery of miR-302/367 to the infarcted myocardium promoted (85). Fibroblast signaling acts as a source of IL-1 positive feedback,
cardiomyocyte proliferation and regeneration by targeting macro- attracting immune cells into the infarct zone (86). Cytokine-activated
phage stimulating 1 (MST1), a central Hippo-YAP player. HOPX is inflammatory fibroblasts modulate the secretion of proteases in-
a key regulator of heart development and induces cardiomyocyte pro- cluding matrix metalloproteinases (MMPs) that are essential for
liferation when overexpressed (14). In a series of studies, the Giacca clearing the infarct of damaged matrix debris (87).
lab determined that miR-590 and miR-199a both promote cell cycle Proliferation
reentry in adult cardiomyocytes by targeting HOPX (70). They sub- The process that has drawn the most focus in fibroblast repair of the
sequently demonstrated that the proproliferative effect of miR-199a heart is their differentiation into myofibroblasts and migration into
is also dependent on its regulation of the Hippo-Yap pathway (71). the infarct zone. Transforming growth factor– (TGF-) is a key
Overexpression of miR-199a using adeno-associated virus 6 in a swine regulator of this phase (88). After MI, TGF-1 is generated by macro-
model of MI resulted in unimpeded cardiomyocyte proliferation that phages producing angiotensin II (AngII). AngII has autocrine func-
progressively reduced the cardiac scar size but eventually led to sudden tion, causing the up-regulation of TGF-1 (89). TGF-1 signaling
cardiac death in 70% of treated animals (72). The loss of FSTL1 has involves a complex cascade of proteins including activating and in-
been shown to be a maladaptive response to injury, whereas its resto- hibitory SMADs, with pleiotropic effects. TGF- suppresses MMPs
ration resulted in increased numbers of proliferating cardiomyocytes (13). (90) while significantly increasing the production of collagens type 1
Recently, Xiao et al. (73) has demonstrated that miR-9 directly inhibits and 3, causing ECM synthesis (91). Activated myofibroblasts express
FSTL1 after MI and that the inhibition of miR-9 results in restored FSTL1, -smooth muscle actin (SMA), allowing for scar contraction.
increases cardiomyocyte proliferation, and preserves cardiac function. Notably, TGF-1 is secreted at an increased rate globally in the
In parallel to targeting these canonical pathways, a set of miRNAs myocardium after injury, not just directly at the site of infarction
prevents proliferation by directly targeting cell cycle regulators such (92), indicating widespread ramifications of MI.
as cyclins. miR-34a and let-7i inhibit cyclin D1 and cyclin D2, Scar maturation
respectively, suppressing their functions through the cell cycle (74, 75). During the maturation phase, ECM proteins secreted by activated
In addition, miR-294 inhibits checkpoint kinase Wee1, preventing fibroblasts are cross-linked to form a stable scar. Over time, the myo-
its suppression of the CDK1/cyclin B1 complex and cell cycle re- fibroblast population in the scar decreases (93) through apoptosis
entry (76). Last, miR-128 inhibits the chromatin modifier SUZ12, and transition into a recently described fibroblast phenotype termed

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 6 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

the matrifibrocyte that is capable of maintaining the scar integrity and harmful miRNAs that induce fibroblast activation. Particular
(94). There is increasing evidence that the myofibroblast phenotype attention has been put on the TGF- signaling pathway (15 of
is reversible in vitro: CFs isolated from patients with heart failure revert 26 studies). Here, we summarize key miRNAs and their mechanisms
to quiescence upon TGF-1 inhibition (95). Therefore, the failure of of action in CFs (Fig. 4A).
myofibroblasts to deactivate has broad implications for clinical worsen- TGF- signaling
ing of MI. Overactivation is driven by multifactorial processes, includ- Many miRNAs regulate parts of the TGF- pathway, including its
ing the persistent secretion of AngII in parts of the heart remote from production, cleavage, and signal transduction. The classical upstream
the site of injury (96). MiRNA therapies targeting fibroblasts seek to regulator of TGF-, c-Fos, is regulated by miR-101a, with overex-
break the positive feedback loop of myofibroblast activation, in which pression of miR-101a leading to reduced fibrosis and improved func-
the stiffening of the heart matrix induces even more TGF- secretion. tion after MI (96). Concurrently, Zhao et al. (97) identified TGF-
receptor 1 (TGFBR1) as another target of miR-101a, using a rat
Recent advances in miRNA therapies targeting model to show decreases in TGF- signal transduction. Another
cardiac fibroblasts potential target for TGF- modulation is AZIN1, as the knockdown
The inhibition of myofibroblast activity is a major focus of ongoing of AZIN1 up-regulated the TGF- expression, with miR-433 direct-

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


research with the identification of beneficial miRNAs that prevent ly reducing AZIN1 in vivo (98). Transfection of miR-433 into CFs
increased fibroblast proliferation and
SMA expression, whereas injection of
A a miR-433 antagomir in rats increased
AZIN1 in post-MI hearts, leading to a
reduction in fibrosis. Proteases cleaving
latent TGF- to its active form can also
be targeted by miRNAs to reduce fibro-
sis. Wang et al. (99) showed that miR-24
inhibits furin, one such protease. In vi-
tro overexpression of miR-24 increased
TGF- secretion and Smad2/3 phos-
phorylation, and miR-24 was found to
be underexpressed in MI tissue.
B Many miRNAs target the cellular
receptors that bind TGF-. Hong et al.
(100) identified TGFBR1 (one of two
main receptors for TGF-) as a target of
miR-22. Overexpression of miR-22 re-
duced AngII activation of CFs in vitro.
Liang et al. (101) described a reciprocal
loop by which TGF- up-regulates
miR-21, which then inhibits TGFBR3, a
negative regulator of TGF- signaling.
The inhibition of TGFBRIII increases
TGF- secretion and Smad3 phosphoryl­
C ation and therefore collagen secretion.
Similarly, Du et al. (102) showed that
miR-328 targets TGFBRIII and that an
injection of anti–mir-328 can improve
cardiac fibrosis post MI in mice.
After binding to the receptor, TGF-
CREDIT: A. KITTERMAN/SCIENCE TRANSLATIONAL MEDICINE

signaling is mediated by decapentaple-


gic (DPP) homologs (SMAD) family.
SMAD2/3 couple to the receptor and
are phosphorylated by TGF- binding,
subsequently binding to SMAD4. SMAD6
Fig. 4. Summary of miRNAs that target fibroblasts, endothelial cells, and immune cells. (A) In CFs, miRNAs can
and SMAD7 inhibit TGF- signaling.
regulate TGF signaling as well as ECM synthesis and pathological proliferation. (B) In ECs, a large number of miRNAs
miRNA mimics, which target SMAD2/3/4
have been shown to regulate angiogenesis, whereas others have been shown to regulate EC apoptosis. (C) In im-
mune cells, miRNAs can regulate multiple aspects of macrophage function including immune cell infiltration, cytokine
and anti-miRs against those that target
production, macrophage polarization, efferocytosis, and phagocytosis, as well as angiogenesis signal release. Select SMAD6/7, can prevent fibrosis. Yuan et al.
well-characterized miRNAs, their targets (written above the arrows), and the processes that they regulate are shown (103) showed the suppression SMAD7
(i.e., miR-24 prevents TGF activation through FURIN signaling). miRNAs prefaced with anti– denote that the inhibi- by miR-21, with maladaptive response
tion of the miRNA is therapeutic (i.e., the inhibition of miR-433 prevents TGF activation through AZIN1 signaling). in fibroblasts. miR-34a is up-regulated
For the complete list of miRNAs reviewed, please see table S1. after MI, and in vivo inhibition of miR-34a

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 7 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

reduced post-MI cardiac fibrosis: SMAD4 was identified as a di- induced collagen mRNA expression in the heart and other organs.
rect target of miR-34a (104). Ischemic exosomes isolated from miR-29 was found to correlate to collagen expression after MI (121),
mouse cardiomyocytes after MI were abundant in miR-92a, which and collagen 1 was identified as a target of miR-133a (122) in
directly targets SMAD7. Overexpression of miR-92a contributed to AngII-induced injury. However, an opposite observation was found
the activation of fibroblasts (105). These post-MI cardiomyocyte with genetic knockout of the miR-29 cluster in mice, causing cardio-
exosomes contain miR-195, which also targets SMAD7 (106). protection and decrease in fibrosis (123). The authors suggest that
Last, the downstream effectors of TGF-, such as connective miR-29 may be more dominantly expressed in cardiomyocytes versus
tissue growth factor (CTGF) (107, 108), can also be targeted by miR- CFs and that whereas miR-29 may be antifibrotic in the fibroblasts,
NAs. Duisters et al. (109) initially identified both miR-133 and miR- it is deleterious in cardiomyocytes, with the knockout effect depen-
30 as inhibitors of CTGF. Later, Chen et al. (110) delivered miR-30a dent on the CM-specific effects.
using adenovirus after MI and observed increased heart function Wingless and Int-1 (WNT) signaling is often dysregulated in fi-
and decreased collagen deposition. brosis (124). miR-199a was shown to be up-regulated during fibroblast
Other pathways activation and to target secreted frizzled-related protein 5 in vitro.
The harmful role of miR-21 in cardiac fibrosis was established by Inhibition of miR-199a reduced fibroblast migration and prolifera-

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


the Thum lab (111, 112), which showed, in a mouse model of heart tion (125). Other studies have found that targeting fibroblast pro-
failure, that miR-21 was dysregulated in fibroblasts but not cardio- liferation is beneficial. Cui et al. (126) showed that miR-574-5p was
myocytes. Inhibition of miR-21 increased the percentage of apop- up-regulated in TGF-–induced fibroblast activation. miR-574-5p
totic fibroblasts in the failing heart. They demonstrated that miR-21 targets ARID3A, which has been implicated in mediating cell cycle
targets Sprouty1, resulting in increased prosurvival extracellular progression. MiR-590 targets ZEB1, an activator of transcrip-
signal–regulated kinase (ERK)/mitogen-activated protein kinase tional regulator CXCR4, and its overexpression inhibited migra-
(MAPK) signaling. Delivery of anti–miR-21 in vivo attenuated fibrosis. tion, proliferation, and collagen secretion in CFs (127).
Similarly, Cardin et al. (113) demonstrated that miR-21 is up-regulated
in atria after MI and contributes to the development of fibrosis and
arrhythmia in atrial fibrillation. miR-21 knockdown reduced atrial ROLE OF miRNAs IN ENDOTHELIAL CELL RESPONSE
fibrosis and stabilized electrical conduction. TO MYOCARDIAL INFARCTION
Yuan et al. (114) identified miR-144 as a PTEN inhibitor in minia- The cardiac muscle has a dense vascular network to meet the high
ture swine. Overexpression of miR-144 or transfection of a PTEN-­ metabolic demands of the tissue, and the continuous EC monolayer
targeting small-interfering RNA (siRNA) in primary human CFs lining serves as a barrier between the blood and myocardium (128).
in vitro increased collagen 1 and SMA mRNA expression. MI severely affects these functions and induces endothelial activa-
Since smooth muscle gene expression is a hallmark of differentiated tion, which facilitates recruitment of inflammatory cells during the
myofibroblasts, miRNAs that regulate smooth muscle phenotype inflammatory phase and mediates the repair and remodeling of the
are potentially therapeutic. miR-143 and miR-145 are transcribed vascular network within the injured cardiac tissue via angiogenesis.
from the same cluster and play a critical role in smooth muscle dif- Thus, miRNA therapy targeting ECs focuses mainly on regulating
ferentiation (115). Li and colleagues (116) showed that miR-143 is inflammatory recruitment and inducing angiogenesis.
up-regulated in human MI tissue samples, and miR-143 inhibitors
reversed the effects of TGF- stimulation on fibroblasts in vitro. Endothelial cell responses to myocardial infarction
miR-143 directly binds to the 3′ untranslated region of Sprouty3, Inflammatory recruitment
activating p38, ERK, and c-Jun N-terminal kinase pathways. In an- Immediately after MI, ischemia activates cardiac ECs to a proin-
other study, Wang et al. (117) showed that miR-145 is sufficient to flammatory and prothrombotic phenotype (129). The ischemic
increase the myofibroblast phenotype of CFs by targeting KLF5, environment causes an increase in the generation of ROS and the
which is a negative regulator of the myocardin serum response factor presence of proinflammatory cytokines, such as TNF and IL-6
(SRF) pathway. miR-145 inhibition in vivo reduced SMA expres- (130). These conditions induce the expression of cell surface adhesion
sion but increased scar size, perhaps indicating a decrease in con- molecules, providing sites of adhesion to facilitate the recruitment
tractile function of the fibroblasts. and attachment of circulating leukocytes. The proinflammatory
The miR-133a family, mir-133a-1, miR-133a-2, and miR-133b, are phenotype results in reduced endothelial nitric oxide (NO) production
also key regulators of the SRF pathway. Of these, the two miR-133a and bioavailability, impairing the ability of the endothelium to regu-
genes are identical and specifically expressed in cardiac myocytes, late vascular permeability and tone in response to external stimuli.
whereas miR-133b is expressed in skeletal muscle (118). miR-133a Although endothelial activation mediates the inflammatory response
is down-regulated in the hearts of patients with MI as well as in within the cardiac tissue, prolonged activation and continued imbalance
animal models (119). miR-133a double knockout mice display severe of ROS and NO generation can lead to permanent adverse effects,
fibrosis and early mortality (118). Expressed specifically in cardio- such as endothelial dysfunction and cellular apoptosis (130).
myocytes, miR-133a appears to act in a paracrine manner, reducing Angiogenesis
the secretion of profibrotic cytokines. Duisters et al. (109) showed Angiogenesis, the process by which new blood vessels form from
that miR-133a regulates the production of CTGF in cardiomyocytes, existing vasculature, is essential for cardiac repair after MI, as it is
a downstream effector of TGF- signaling. needed to restore sufficient blood flow to the injured tissue (131).
ECM proteins can also be targeted as the end product of myo- Revascularization occurs at the site of the infarct as well as throughout
fibroblast activation. The Olson lab demonstrated that miR-29 is the surrounding injured tissue via proliferation and migration of
decreased after MI and that it targets ECM protein mRNAs, including ECs. Ischemia induces expression of growth factors, which promote
collagens, fibrillin, and elastins (120). Inhibition of miR-29 in vivo EC survival, migration, and proliferation, such as the vascular

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 8 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

endothelial growth factor (VEGF) and fibroblast growth factor MAPK pathway, the proangiogenic miR-126 and miR-132 both
(FGF) families (132). The hypoxic environment created during suppress negative regulators of MAPK activity, Spred-1 and RASA1,
ischemia up-regulates the expression of hypoxia-inducible transcrip- respectively, whereas anti-angiogenic miR-24 targets p21-activated
tion factors (HIFs), such as HIF-1, which activate the angio- kinase (PAK4), inhibiting MAPK activity (138–141). miR-24 also
genic process. targets the eNOS signaling pathway, along with miR-199a and miR-155,
all of which suppress migration, proliferation, and tube formation
Recent advances in miRNA therapies targeting of ECs (142, 143). Inhibition of these miRNAs increases NO bio-
endothelial cells availability and promotes angiogenesis in these studies. Although
Of the studies reviewed, relatively few focused on the EC population these miRNAs target molecules of pathways activated by the binding
(24 of 213). Although some studies focus on preventing EC apoptosis of VEGF, certain miRNAs target VEGF directly, such as the antian-
or address other effects (5 of 24), most studies target the process of giogenic miR-590 (144).
angiogenesis (19 of 24). Here, we summarize key miRNAs and their Several other miRNAs affect angiogenesis by modulating signal-
mechanisms of action in ECs (Fig. 4B). ing pathways that are not VEGF associated. In two separate studies,
Apoptosis miR-210 was shown to promote angiogenesis, with overexpression

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Apoptosis of ECs disrupts the barrier function of the endothelium increasing EC proliferation and migration by up-regulating hepato-
and impairs its ability to effectively regulate the inflammatory re- cyte growth factor expression and targeting focal adhesion kinase
sponse after injury. miRNA involvement in EC apoptosis has been (145, 146). miR-92 also showed, in two separate studies, that it targets
primarily studied within the context of atherosclerotic models, the proangiogenic integrin 5 (ITGF5) and that its inhibition sub-
which is a major cause of complications such as MI. Here, beneficial stantially increased angiogenesis and granulation tissue formation
miRNAs prevent, whereas harmful ones promote EC apoptosis. (147, 148). Because miR-26a targets SMAD1, its inhibition led to
The atherogenic factor oxidative low-density lipoprotein (ox-LDL) increased angiogenic activity and reduced infarct size in a murine
induces EC dysfunction and apoptosis and has been used both model of MI. In contrast, miR-27b promotes vascularization by
in vitro and in vivo to identify and investigate associated miRNAs. targeting notch ligand DLL4 (149). Last, miR-185 inhibits prolifer-
miR-26a was identified as dysregulated during atherosclerosis: Its ex- ation, migration, and tube formation by targeting the CatK gene
pression was suppressed in a dose-dependent manner in human aortic (150). Inhibition of miR-185 significantly promoted these functions
ECs (HAECs) treated with ox-LDL (133). Overexpression of miR-26a of angiogenesis under hypoxic conditions.
targeted and repressed activity of transient receptor potential cation Other effects
channel subfamily C member 6 (TRPC6), a calcium-permeable channel Bayoumi et al. (151) determined that miR-532 overexpression de-
subunit, inhibiting apoptosis by inhibiting cytosolic calcium over- creased endothelial-to-mesenchymal transition (EndMT) in cardiac
load, which triggers the calcium activated apoptotic pathway. ECs. miR-532, which is up-regulated by 2 adrenergic receptor/
In a separate study, ox-LDL–induced apoptosis was investigated in -arrestin activity, exerts this cardioprotective function by targeting
HAECs and human umbilical vein endothelial cells (HUVECS), and protease serine 23 (PRSS23), a positive regulator of maladaptive
miR-150 was up-regulated during the process (134). Overexpression EndMT. The metabolic state of ECs can also alter MI outcomes, as
of miR-150 enhanced cellular apoptosis, whereas inhibition of miR-150 Bartman et al. (152) identified that miR-21 increased glycolytic ac-
alleviated apoptosis. The proapoptotic miR-150 negatively regulates tivity in ECs in hypoxic conditions. miR-21 was up-regulated under
expression of Cu/Zn superoxide dismutase (SOD1) through the direct hypoxic conditions only in ECs, not in cardiomyocytes or fibroblasts,
targeting of transcription factor ELK1. Last, miR-324 was shown to despite being predominantly expressed in fibroblasts under normoxic
protect against hypoxia-induced apoptosis in endothelial progenitor conditions. Inhibition of miR-21 resulted in larger infarct sizes in a
cells (EPCs), which are active participants in the recovery process myocardial IRI murine model.
after MI, supporting both vascular endothelium repair and angio-
genesis (135). Increasing expression of miR-324 down-regulates the
MTFR1 gene, which, in turn, reduces mitochondrial fragmentation, ROLE OF miRNAs IN IMMUNE CELL RESPONSE
stabilizes mitochondrial membrane potential, and results in decreased TO MYOCARDIAL INFARCTION
cellular apoptosis in hypoxic conditions. MI triggers an inflammatory cascade in the heart starting with an
Angiogenesis acute proinflammatory response, which is gradually replaced by
Angiogenesis is a tightly regulated process involving multiple mo- an anti-inflammatory reparative phase. Therapeutic advancements
lecular pathways. miRNAs modulate many of the pathways involved, generally aim to dampen the initial inflammatory response and
with beneficial miRNAs promoting and harmful miRNAs impeding promote the proreparative phase.
the angiogenic process.
Multiple miRNAs that regulate angiogenesis modulate pathways Inflammatory responses to myocardial infarction
activated by binding of VEGF (specifically VEGFA) to cell surface Activation of inflammation
receptors, including HIF-1, MAPK, and endothelial nitric oxide Upon injury, resident immune cells located in perivascular areas are
synthase (eNOS), which all contribute to promoting angiogenesis activated, resulting in an acute inflammatory response. In the early
(132). Modulating HIF-1a is the proangiogenic miR-424, which is inflammatory phase, the infiltration of immune cells in the infarct area
up-regulated in ECs under hypoxic conditions and targets Cullin 2 and accumulation of proinflammatory chemokines/cytokines result
(136) to stabilize and up-regulate HIF-1. In contrast, the harmful in not only the clearance of cellular debris but also in further damage
miR-223, although also up-regulated in ischemic conditions, nega- to cardiac tissue. Macrophages, among the largest resident cell popu-
tively regulates HIF-1 signaling by targeting ribosomal protein S6 lations in cardiac tissue, are predominantly responsible for the pro-
kinase B1 and inhibits EC migration and proliferation (137). In the duction of inflammatory cytokines post MI. They release IL-1, IL-6,

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 9 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

and TNF, triggering further responses from multiple cell types, knockdown of miR-375 all suppressed CD68+ cell accumulation in
both locally and remotely. The activation of this inflammatory cascade the infarct border zone, leading to improved cardiac function with
mediates remodeling throughout the myocardium. decreased infarct size (158–161).
Resolution of inflammation Inflammatory cytokine production
Increasing evidence indicates that the initial inflammatory response The accumulation of proinflammatory cytokines plays a critical role
elicited by MI induces resident macrophages to switch from an in governing the progression and extent of tissue remodeling. The
inflammatory M1 phenotype to a resolving M2 phenotype. M2 inhibition of this process by miRNA therapies successfully decreases
macrophages secrete anti-inflammatory signals critical for the re- myocardial infarct size and improves cardiac function. miR-21
pair response, allowing wound healing and scar formation, thereby mimic delivered to monocytes/macrophages in mice reduced in-
limiting infarct size. Failure to activate reparative responses leads flammatory cytokine expression by directly inhibiting KBTBD7-­
to adverse cardiac remodeling with further damage and, ultimately, mediated DAMP (damage-associated molecular patterns)–triggered
heart failure. inflammatory responses in macrophages (154). The delivery of
miR-106b, miR-148b, and miR-204 individually encapsulated into
Recent advances in miRNA therapies targeting immune cells polyketal nanoparticles in macrophages decreased Nox2 expression,

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Of the studies surveyed, 17 of 213 focus on immune cells. Although inhibiting superoxide production and expression of proinflammatory
some studies have applied miRNA therapies to either reduce the genes IL-1, IL-6, and TNF- in vitro (158). Anti–miR-375 injection
damage of the inflammatory response (10 of 17 studies) or focus on after MI in mice decreased inflammatory response by reducing the
promoting the reparatory pathways (3 of 17 studies), several other production of several proinflammatory cytokines including IL-6,
effective therapies have targeted both responses, exerting the overall interferon gamma-induced protein 10 (IP-10), monocyte chemoattrac-
functional benefits as an ensemble (4 of 17 studies). In targeting tant protein-1 (MCP-1), macrophage inflammatory protein-1 alpha
immune cells, beneficial miRNAs aim to prevent infiltration of im- (MIP-1), regulated on activation, normal T cell expressed and secreted
mune cells and inflammatory signal production in the infarct area and (RANTES), and MIP1- (160). After reperfusion in rats and swine,
promote M2 macrophage polarization, efferocytosis, phagocytosis, intramyocardial injection of miR-181b–enriched CDC-exo also
and angiogenesis signal release. Here, we summarize key miRNAs attenuated the expression of proinflammatory genes by targeting
and their mechanisms of action in immune cells (Fig. 4C). PKC (159).
Infiltration M2 macrophage polarization
Infiltration of immune cells such as macrophages and neutrophils Polarization of macrophages toward the anti-inflammatory M2
into the infarct area is one of the initial inflammatory responses after phenotype has been found to confer beneficial effects on cardiac
MI. Many miRNAs have been shown to regulate this process, pro- repair. Intramyocardial injection of MSC-exo containing miR-182
viding interesting therapeutic targets. Recently, miR-141 has been after myocardial IRI reduced infarct size and alleviated damaged
shown to suppress neutrophil/leukocyte adhesion to ECs by modu- myocardial inflammation in mice. Mechanistically, miR-182 polarized
lating expression of the cell adhesion associated protein intercellular macrophages toward the M2 phenotype and regulated downstream
adhesion molecule–1, which plays a crucial role in regulating the Toll-like receptor 4 (TLR4) expression, mitigating proinflammatory
migration of leukocytes across the endothelium into the myocardium. cascades and enhancing subsequent repair (162).
In vivo, pretreating mice with intravenous miR-141 mimics before Tail vein injection of miR-21 mimic delivered to cardiac macro-
IRI successfully reduced CD11b+ myeloid cells and F4/80+ macro- phages by encapsulated nanoparticles promoted macrophage switch-
phages accumulation in the ischemic myocardium (153). ing from a proinflammatory to reparative phenotype, thus inducing
miR-21 knockout mice showed enhanced infiltration of CD11b+ the resolution of inflammation (163). The polarization state was
monocytes/macrophages in myocardium, whereas miR-21 overex- also shifted in cardiac macrophages isolated from CDC-exo–treated
pression markedly inhibited it (154). However, contrasting findings rats and swine. miR-181b inhibition of PKC expression further
in another study showed that miR-21 inhibition in cardiac allografts demonstrates that PKC acts as a downstream effector of CDC-exo–
led to reduced infiltration of CD45+ leukocytes, but there was no mediated cardioprotection (159). Moreover, increased CD206+ cells
difference observed for macrophage infiltration (155). Intravenous (M2 macrophage marker) in the left ventricular infarct border zone
injection of miR-144 mimics resulted in reduced infiltration of macro- of anti–miR-375–treated mouse hearts after MI indicate that miR-
phages in the border zone, decreased MMP-mediated inflammation, 375 knockdown can trigger a macrophage switch toward the anti-­
and reduced fibrosis and apoptosis. The beneficial effects of miR-144 inflammatory M2 phenotype (160). In vitro studies in macrophages
therapy were associated with mTOR- and P62-mediated autophagy further demonstrated that miR-375 modulated AKT signaling through
signaling with improved cardiomyocyte survival. Alterations in phosphoinositide-dependent kinase-1 (PDK-1). PDK-1 knockdown
TGF- signaling also demonstrated a role for miR-144 in modulat- abolished the macrophage polarization effect of miR-375, further
ing the local inflammatory response (156). confirming that PDK-1 is a critical mediator in miR-375–regulated
Intramyocardial injection of anti–miR-224–expressing lentivirus macrophage phenotype shift.
elevated vascular cell adhesion molecule–1 expression, regulating Targeting efferocytosis and phagocytosis
inflammation-associated vascular adhesion and transendothelial The clearance of apoptotic cells by macrophages is thought to play
migration of macrophages/leukocytes, and MMP-2, an inflammatory a critical role in the reparative phase allowing for further recovery
mediator participating tissue remodeling in pathological processes after MI. MI-associated transcript (MIAT), a long noncoding RNA,
(157). Other studies have shown that the overexpression of miR-106b, has been shown to regulate phagocytosis by acting as a sponge of
miR-148b, and miR-204 injection of miR-181b–enriched cardiosphere-­ miR-149, which directly targets antiphagocytic molecule CD47 (164).
derived exosomes (CDC-exo) or miR-24–enriched umbilical Knockdown of MIAT or miR-149 overexpression in murine macro-
mesenchymal stem cells (MSCs) secreted exosomes (MSC-exo), and phages reduced CD47 expression and promoted efferocytosis and

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 10 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

phagocytosis, which consequently increased resolution of inflam- versus cardiomyocytes. Another extensively studied miRNA that
mation and clearance of apoptotic cells by macrophages. Similarly, falls into this category is miR-199a. miR-199a was shown to induce
transfecting EPCs derived from the peripheral blood of patients adult cardiomyocyte proliferation after MI (70), and subsequent
with ST-segment elevation MI with miR-324 mimic resulted in re- studies identified multiple proproliferative pathways regulated by
duced apoptosis, increased proliferation, and elevated phagocytosis miR-199a in cardiomyocytes (71). Additional studies have demon-
by targeting and inhibiting MtFr1 after peroxide-induced oxidative strated that miR-199a can regulate other crucial pathways in cardio-
stress (135). The protective role of miR-324 overexpression against myocytes including glucose metabolism, hypertrophy, apoptosis,
oxidative stress–induced EPCs injury suggests that therapies lead- and autophagy with divergent effects (66, 170–173) and negatively
ing to correction of miRNA expression may contribute to tolerance affect angiogenesis in ECs as well as induce pathological fibrosis in
against injury by regulating phagocytosis pathways. fibroblasts (125, 143).
Targeting angiogenesis signal release Another noteworthy miRNA is miR-21. Multiple studies have
Macrophages engage in cross-talk with other cell types and release a demonstrated that miR-21 prevents cardiomyocyte apoptosis after
variety of angiogenic signals to initiate and regulate angiogenesis in ischemic injury through direct inhibition of the proapoptotic mole-
ECs in response to MI. After MI, M1 macrophages secrete proin- cule PDCD4 (38–40). In addition, treatment with miR-21 mimics

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


flammatory exosomes (M1-exo), which contain abundant miR-155 has been beneficial to ECs by preventing apoptosis (152) and to im-
(142). M1-exo containing miR-155 suppressed expression of several mune cells by preventing inflammatory activation (152, 154, 163).
genes, including RAC1, PAK2, Sirt1, and AMPK2, and modulated However, despite consensus on the beneficial effects of miR-21 in
Sirt1/AMPK2-eNOS and Rac family small GTPase 1 (RAC1) - p21 cardiomyocytes and ECs, multiple studies have demonstrated harmful
(RAC1)–activated kinase 2 (PAK2) signaling pathways, leading to the effects of miR-21 in fibroblasts, and other studies have shown con-
inhibition of angiogenesis and cardiac dysfunction. The antiangio- tradictory effects in immune cells. The knockdown of miR-21 pre-
genic function of miR-155 makes its inhibition an attractive thera- vents pathological fibrosis by targeting the ERK/MAPK and TGF-
peutic strategy for MI (142). pathways (101, 111–113). In immune cells, miR-21 has been shown
to both prevent macrophage infiltration and induce leukocyte infil-
tration (154, 155). Ultimately, despite contradictory effects, the stron-
miRNAs WITH PLEIOTROPIC EFFECTS gest evidence for the therapeutic capabilities of miR-21 comes from
Most miRNA studies focus on one miRNA in a singular cell type. a recent study by Hinkel et al. (174), which demonstrated that inhibi-
However, the literature demonstrates that many miRNAs have tion of miR-21 prevented myocardial dysfunction in a pig model of MI.
pleiotropic effects on myocardial recovery after MI, spanning dif- Multiple miRNAs have been shown to have divergent effects on
ferent pathways, processes, and cell types. Some pleiotropic miRNAs fibroblasts compared to cardiomyocytes. For example, miR-22,
regulate many points along the same pathway, whereas others regu- miR-29, miR-101, and miR-155 are all beneficial to fibroblasts by
late different pathways. Interestingly, multiple of these miRNAs preventing activation and proliferation while being harmful to cardio-
demonstrate divergent effects among cell types—promoting recovery myocytes by promoting apoptosis or autophagy (59, 96, 97, 100, 120,
processes in some cell types while impeding recovery processes in 142, 175–179). In contrast, miR-144 prevents cardiomyocyte apop-
other cell types. Developing a clear mechanistic understanding of tosis (beneficial) while also promoting fibroblast proliferation
the pleiotropic effects of miRNAs will greatly advance their clinical (harmful), by targeting the same PI3k/AKT pathway in both cell
translation. types (156). Collectively, these miRNAs demonstrate the complexity
of miRNA effects across multiple cell types in the heart and suggest
miRNAs with convergent effects the difficulties with using nontargeted miRNA therapies. Thus, a
Some miRNAs have convergent effects: promoting or impeding re- cardiomyocyte-centric approach to miRNA therapy limits our un-
covery processes in all cell types. For example, miR-210, miR-324, derstanding of pleiotropic effects.
miR-494, and miR-532 all have beneficial effects on both cardiomyo-
cytes and ECs (42, 135, 145, 146, 151, 165–169). Although miR-324
had the same target (MTFR1) and effect (prevention of apoptosis) PROSPECTS AND CHALLENGES FOR CLINICAL TRANSLATION
in both cell types (135), the other three miRNAs had different tar- Promising preclinical studies have spurred interest in clinical trans-
gets and affected different pathways in each cell type. In separate lation of miRNA therapies. Currently, there are no miRNA thera-
studies, miR-24 was shown to have beneficial effects on cardiomyo- pies with U.S. Food and Drug Administration (FDA) approval;
cytes, fibroblasts, and ECs through the prevention of apoptosis, however, several early-stage biotechnology companies are focused
prevention of fibrosis, and activation of angiogenesis, respectively solely on miRNAs, such as Miragen Therapeutics, Cardior Thera-
(24, 99, 141). On the other hand, miR-92a has been shown to have peutics, and Regulus Therapeutics. One phase 1 trial is currently
harmful effects on fibroblasts and ECs through the activation of ongoing to assess the safety of a miR-132 inhibitor (NCT04045405)
fibrosis and the prevention of angiogenesis (105, 147, 148). Because in ischemic heart disease. There are several clinical trials for treat-
of their convergent effects on multiple cell types studied, these ment of related pathologies, including the use of a miR-21 inhibitor
miRNAs may be particularly promising therapeutic targets. for prevention of renal fibrosis (NCT03373786) and a miR-29 mimic
for pulmonary fibrosis (NCT03601052).
miRNAs with divergent effects As discussed here, mimicry of “beneficial” miRNAs or inhibition
Interestingly, most of the pleiotropic miRNAs identified demonstrate of “harmful” miRNAs both have therapeutic potential. However, the
divergent effects: promoting recovery processes in some cell types latter has benefited from past innovation in the RNA interference
while impeding recovery processes in other cell types. We already field, which has recently begun, achieving success in the clinic, with
described miR-29, which demonstrated contradictory effects on CFs the first drug of its class, Patisiran, approved by the FDA in 2018. It

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 11 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

is thus not surprising that of the current miRNA therapeutics in MI. Notably, they also show that intravenous infusions were as
either phase 1 or 2 clinical trials, the majority are ASOs targeting a effective as IC injections, suggesting the possibility for minimally
specific miRNA (180), although mimics are also being tested. Similar invasive delivery of miRNA therapies.
to the siRNA field, these burgeoning new therapies face challenges Intravenously injected ASOs delivered without a vehicle still re-
including considerations for therapeutic timing, optimal oligo- quire large doses to be effective in vivo (185). In addition, efficient
nucleotide optimization, in vivo delivery, and side effects. delivery to the target tissue—in this case, the heart—is essential for
optimizing in vivo delivery of miRNA therapies. For this purpose,
Oligonucleotide modifications carriers such as lipid nanoparticles (LNP) and nanoparticles (193)
Naked oligonucleotides rapidly accumulate in the kidney and liver have been explored. Traditional LNPs have been clinically validated
and are quickly cleared from the circulation (181). Moreover, cleavage for siRNA delivery, including Patisiran. However, their use comes
by serum exonucleases and degradation in the intracellular endosomal at the cost of additional components and toxicities. Because these
compartment reduces drug potency. To improve stability, various systems have been reviewed extensively (181, 194), they will not be
chemical modifications to the nucleic acid backbone have been used discussed in detail here.
in the design of miRNA mimics and inhibitors. Extracellular vesicles, including exosomes, are plasma membrane–

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Phosphothiorates substitute a sulfur for oxygen in the phosphate bound cell-secreted vesicles that transfer biologically active cargo
group of the nucleotide. Compared to unmodified oligonucleotides, between cells and act as the endogenous analog of LNPs. In the field
phosphothiorates are more resistant to nucleases and thus drastically of heart repair, exosomes emerged first as paracrine mediators of
increase circulation time (182). However, compared to an unmodified cell therapy. They are secreted by all cells and contain a variety of
phosphodiester bond, they have decreased binding affinity to their contents, including miRNAs (195). Exosomes from several cell types,
target as measured by a lower melting temperature (Tm) (183). To including MSCs, cardiac progenitor cells, and induced pluripotent
address these issues, second-generation designs centered around stem cell–derived cardiomyocytes (iPS-CMs), are effective in pro-
the use of 2′-O modifications of the ribose sugar and locked nucleic moting cardiac repair (196–198). Several advances have been made
acid (LNA) modifications while reducing and interspersing the num- toward clinical translation, including the development of Good
ber of phosphothiorate modifications. 2′-O-methylation (2′OMe), Manufacturing Practice protocols for the collection of therapeutic
first tested in the early 2000s, enhances target binding (184). exosomes (199) as well as toward altering their cargo. Unlike trans-
Krützfeldt et al. (185) used 2′OMe oligonucleotides with spaced planted cells and synthetic LNPs, autologous exosomes do not trigger
phosphothiorate bonds and a cholesterol group at the 3′ end to in- the immune system. However, barriers to their use remain, like
crease resistance to exonucleases while preserving nuclease activity, addressing their heterogeneity and production. Eventually, highly
so termed “antagomirs” (186). defined engineered exosomes may represent a way to fine-tune
LNA modifications contain methylene linkages of the 2′O to the miRNA therapy for the heart. For example, MSCs overexpressing
4′C in the sugar backbone, locking the structure into a C3′-endo miR-133 were more effective in cardioprotection than control vector
sugar conformation (186). These afford an increase in Tm while MSCs in a rat model of MI, an effect attributed to an increase in
simultaneously increasing nuclease resistance (187). Given their high therapeutic exosome content (200).
binding affinity, much shorter LNA oligonucleotides can be de-
signed. This has both implications for target specificity, as shorter Tissue-specific delivery
sequences more likely bind multiple families of miRNAs, and At this time, targeted delivery of miRNA therapeutics to the heart
the related cost of synthesizing such an oligonucleotide. Recently, remains an unmet need. Biodistribution studies show that LNP and
Obad et al. (188) reported the use of an 8-mer LNA oligo capable of exosomes are entrapped by the liver and spleen and filtered from
powerful anti-miRNA effects. the blood by the kidneys’ glomerular barrier due to their size (201).
Last, miRNA sponges have been explored as alternatives to ASOs With only an estimated 2000 to 5000 copies of oligonucleotide re-
(189). These dominant-negative inhibitors contain multiple target quired within the cell for gene knockdown, drug loading doses can
sites complementary to a specific miRNA seed sequence. They have be reduced if delivery can be constrained to the heart (202). The
several advantages over traditional oligonucleotides, including the conjugation of ASOs to N-acetylgalactosamine (GalNAc) to specif-
ability to silence families of RNA sharing a common seed. However, ically target asialoglycoprotein receptors in the liver has contributed
further progress on their design and delivery is required for clinical greatly to the commercial success and translation of Patisiran (203).
translation, and this area of research still trails ASOs in terms of Identifying similar extrahepatic targets, including in the heart, has
optimization (190). not yet been achieved and is a key focus.
To this end, homing cardiac targeting peptides and engineered
Delivery vehicles exosomes have been shown to increase exosome and miRNA deliv-
The route of administration of miRNA therapeutics greatly alters ery in cardiomyocytes after IC injection (204). Chemical aptamers
their potency. Local administration methods such as intracoronary can alter biodistribution. Xue et al. (205) used a nanoparticle den-
injection (IC), hydrogel-based patches, and intramyocardial injec- drimer approach to target miR-1 inhibitors to the AngII receptor
tion have been studied (table S1). The availability of percutaneous 1 in post-MI hearts. Antibody-based targeting can also alter cardiac
coronary intervention as the preferred method for revascularization homing. Immunoglobulins are immunologically privileged proteins
after MI (191) makes IC injection perhaps the most effective local and are continually recycled in the circulation. Conjugation of siRNAs
delivery method. Despite this, the multiple dosing of locally admin- to an anti-CD71 Fab′ fragment allowed for durable gene silencing
istered drugs to the heart is invasive and difficult in humans. In a in cardiac tissues over a month’s time (206). Liu et al. (207) used
preclinical study of MI in 135 swine, Foinquinos et al. (192) demon- anticardiac troponin antibodies to target liposomes containing anti–
strated the efficacy of IC injections of miR-132 immediately after miR-1 to ischemic myocardium. However, the addition of a large

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 12 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

macromolecule may also lead to several disadvantages. Genentech’s linked to carcinogenesis and metastasis in certain cancers, such as
THIOMAB antibody-siRNA conjugates demonstrated the ability to melanoma and gastric cancer (216, 217). Similarly, inhibition of
specifically target prostate cancer in vivo but exhibited suboptimal miR-34a has been shown to improve cardiac remodeling after injury
gene silencing due to sequestering in the endocytic department (208). (74, 218), although, at the same time, miR-34a has been studied as a
In addition, targeting specific receptors may have other effects tumor suppressor, and its mimicry has demonstrated anticancer
besides delivering the therapeutic molecule, such as inhibition or effects (219, 220). Collectively, these examples further highlight a
activation of its own downstream signaling pathway, which adds need to understand cell type–specific effects of miRNAs as well as
complexity to this delivery modality. the need for tissue-specific delivery.

Endosomal escape Alternative preclinical models


After organ-specific targeting, there remains a barrier to entry at Another challenge in miRNA therapeutic development, similar to
the cellular level. miR-mimics and anti-miRs are relatively large genome-based therapies, is the lack of adequate drug-testing
(~10,000 kDa) and highly negatively charged, preventing membrane platforms. Genomic differences in nonprimate animal models limit
transport. Naked nucleic acid or those bound in carriers are ubiqui- their use as a predictive model of the effects of a specific miR-mimic

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


tously taken up by endocytosis (209). Endocytosed molecules then or anti-miR. Moreover, interspecies differences in ion channels and
enter the endosomal system and eventually are degraded in the biological pathways fail to recapitulate human biology; for example,
lysosome (210). To have a clinical effect, miRNA therapeutics must the mouse heart beats nine times faster than the human heart (221).
escape the endosomal pathway into the cytosol. Using traceable Current cardiovascular drug testing relies heavily on simplified
siRNAs, Gilleron et al. (211) estimated that just 1% of delivered models such as human embryonic kidney 293 cells and Chinese
oligonucleotides actually escape the endosomal pathway. The exact hamster ovary cells overexpressing ion channels (222). Of the studies
biological mechanisms of how escape occurs remain to be elucidated. surveyed here, most used primary rat cardiomyocytes or the rat
Approaches to designing delivery carriers, which bypass the endo- H9C2 cell line, highlighting a need for in vitro human disease models.
somal system via diffusion through the endosomal membrane or Although primary human cardiomyocytes would be an ideal candi-
disrupting the compartment pH, have been explored (212). Solving date for such a model, they are in prohibitively short supply and
the endosomal escape barrier will be critical for miRNA therapeutic difficult to isolate. Recently, iPS-CMs have emerged as a promising
development moving forward. source to fill this void (223). iPS-CMs can be produced in near lim-
itless quantity and matured to adult-like phenotypes (224), allowing
Side effects of miRNA therapies for high-throughput screening of thousands of miRNA candidates.
Like any drug, miRNA therapeutics are not without safety concerns. The generation of more complex, three-dimensional models of hu-
For example, phosphothiorates have also been reported to react man cardiac tissues using iPS-CMs can further be used to model
with proteins, including FGF2 (213). Although careful sequence MI (225). Engineered cardiac tissue can also help shed light on miRNA
selection can eliminate off-target RNA hybridization, many oligo- transfer between cardiac cell types and myocyte-nonmyocyte com-
nucleotide modifications produce nonspecific effects, including munication, through extracellular vesicles or otherwise (226). Elu-
differences in protein expression when compared to an unmodified cidating this biology may help further the design and development
siRNA (214). of miRNA therapeutics. How closely these models recapitulate
Nucleic acids also broadly trigger the immune system, activating human biology remains to be seen (227); however, they represent
the TLR family (215). The use of delivery vehicles such as LNPs, exciting technologies that can potentially decrease drug devel-
which are designed to shield ASOs from the immune system, opment cost.
adds further reagents that may trigger an inflammatory response. A
miRNA-34a mimic was halted in phase 1 (NCT02862145) studies Key remaining questions
after several patients developed severe adverse immune reactions. There are several questions, which, when elucidated, would greatly
Development of these therapeutics can be thus unpredictable. advance the field of miRNA therapy for MI. A key question that
Abrogating the acute response to MI may also be dangerous. remains is the timing of therapeutic delivery. The processes of cardio-
Therapies may be able to salvage damaged but viable cells adjacent myocyte death, fibroblast activation, and immune cell response
to the infarct; however, an injured cardiomyocyte that is rescued may occur acutely after ischemia onset. Because of the acute nature of
have impaired function, leading to arrhythmia. Similarly, inter- cardiac injury, is it then necessary for intervention to take place rapid-
ference of fibroblast function after MI has long been controversial, ly after symptom onset? Or can a treatment administered in the later
due to fear of cardiac rupture after failure to generate a scar. Al- stages of wound healing (3 to 4 weeks) still show clinical improve-
though no studies reviewed here reported such outcomes, developing ment? Along a similar vein, could miRNA therapies potentially be
clear understandings of the mechanisms underlying miRNA thera- preventative, suppressing these maladaptive processes before they
pies will go a long way toward preventing these dangerous side begin? Very few preclinical studies have examined the timing of
effects as we continue to search for more potent miRNA therapies. these deliveries and instead apply both the insult and the treatment
The ability of miRNAs to regulate a variety of processes in differ- simultaneously. Furthermore, the potential duration of clinical ben-
ent cell types leads to the additional concern of perturbations in efit from miRNA therapy is not well defined. Given the short-acting
signaling outside of the target organ. Although promoting cardio- nature of miRNAs, it seems that repeated dosing would be required
myocyte proliferation may be desirable for cardiac recovery, altering for long-lasting clinical benefit if early intervention is not possible.
those same pathways in other organs may result in tumorigenesis. Should we target one cell type’s pathobiology or multiple? Is there a
The aforementioned miR-199a, which caused uncontrolled cardio- dominant effect of manipulating one cell type over another? The
myocyte proliferation and subsequent arrhythmia, has also been pleiotropic nature of miRNAs with convergent and divergent effects

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 13 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

is one of its key advantages, yet it adds a degree of complexity to 2. S. A. Hunt, Taking heart—cardiac transplantation past, present, and future. N. Engl.
miRNA drug discovery unparalleled in other treatment modalities. J. Med. 355, 231–235 (2006).
3. S. D. Prabhu, N. G. Frangogiannis, The biological basis for cardiac repair after myocardial
The recent advancements in human iPS-derived in vitro models,
infarction: From inflammation to fibrosis. Circ. Res. 119, 91–112 (2016).
including the ability to generate complex, three-dimensional models 4. M. Ghildiyal, P. D. Zamore, Small silencing RNAs: An expanding universe. Nat. Rev. Genet.
of human cardiac tissues, may represent a powerful and efficient 10, 94–108 (2009).
strategy in answering some of these challenging questions. 5. D. P. Bartel, MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116,
281–297 (2004).
6. E. N. Olson, MicroRNAs as therapeutic targets and biomarkers of cardiovascular disease.
Sci. Transl. Med. 6, 239ps3 (2014).
CONCLUSION
7. A. E. Frampton, L. Castellano, T. Colombo, E. Giovannetti, J. Krell, J. Jacob, L. Pellegrino,
Current interventions fail to recover the injured myocardium after L. Roca-Alonso, N. Funel, T. M. H. Gall, A. De Giorgio, F. G. Pinho, V. Fulci, D. J. Britton,
MI, requiring the development of previously unexplored cardiopro- R. Ahmad, N. A. Habib, R. C. Coombes, V. Harding, T. Knösel, J. Stebbing, L. R. Jiao,
tective strategies. Here, we reviewed miRNAs as powerful regulators MicroRNAs cooperatively inhibit a network of tumor suppressor genes to promote
involved in post-MI remodeling and as the potential targets for MI pancreatic tumor growth and progression. Gastroenterology 146, 268–277.e18 (2014).
8. D. M. Yellon, D. J. Hausenloy, Myocardial reperfusion injury. N. Engl. J. Med. 357, 1121
therapy. Focusing on literature within the past 3 years and the founda- (2007).

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


tional studies within the past 10 years, we identified a total of 213 9. S. Elmore, Apoptosis: A review of programmed cell death. Toxicol. Pathol. 35, 495–516
publications describing the modulation of miRNAs in the treatment (2007).
of MI, involving 116 unique miRNAs. Most of these miRNAs have 10. D. A. Kubli, Å. B. Gustafsson, Cardiomyocyte health: Adapting to metabolic changes
been shown to play both beneficial and harmful roles in the pro- through autophagy. Trends Endocrinol. Metab. 25, 156–164 (2014).
11. S. E. Senyo, M. L. Steinhauser, C. L. Pizzimenti, V. K. Yang, L. Cai, M. Wang, T.-D. Wu,
gression of MI with divergent effects on different target cell types.
J.-L. Guerquin-Kern, C. P. Lechene, R. T. Lee, Mammalian heart renewal by pre-existing
To successfully translate the miRNA-based therapies of MI to the cardiomyocytes. Nature 493, 433–436 (2013).
clinic, a more detailed understanding of the mechanisms and dynamics 12. J. Wang, S. Liu, T. Heallen, J. F. Martin, The Hippo pathway in the heart: Pivotal roles
of miRNA effects on each cell type needs to be clearly defined. Clin- in development, disease, and regeneration. Nat. Rev. Cardiol. 15, 672–684 (2018).
ical translation of these therapies will also require improved admin- 13. K. Wei, V. Serpooshan, C. Hurtado, M. Diez-Cuñado, M. Zhao, S. Maruyama, W. Zhu,
G. Fajardo, M. Noseda, K. Nakamura, X. Tian, Q. Liu, A. Wang, Y. Matsuura, P. Bushway,
istration protocols, organ-specific delivery, and more predictive
W. Cai, A. Savchenko, M. Mahmoudi, M. D. Schneider, M. J. B. van den Hoff, M. J. Butte,
preclinical models. P. C. Yang, K. Walsh, B. Zhou, D. Bernstein, M. Mercola, P. Ruiz-Lozano, Epicardial FSTL1
reconstitution regenerates the adult mammalian heart. Nature 525, 479–485 (2015).
14. Y. Liu, W. Zhang, The role of HOPX in normal tissues and tumor progression. Biosci. Rep.
SUPPLEMENTARY MATERIALS 40, BSR20191953 (2020).
stm.sciencemag.org/cgi/content/full/13/580/eabd0914/DC1
15. D. A. Morrow, Myocardial Infarction: A Companion to Braunwald’s Heart Disease (Elsevier, 2016).
Table S1. Complete list of miRNAs reviewed.
16. J.-X. Wang, J.-Q. Jiao, Q. Li, B. Long, K. Wang, J.-P. Liu, Y.-R. Li, P.-F. Li, MiR-499 regulates
Table S2. Summary of miRNAs reviewed.
mitochondrial dynamics by targeting calcineurin and dynamin-related protein-1.
References (228–298)
Nat. Med. 17, 71–78 (2011).
17. K. Wang, T.-Y. Gan, N. Li, C.-Y. Liu, L.-Y. Zhou, J.-N. Gao, C. Chen, K.-W. Yan, M. Ponnusamy,
Y.-H. Zhang, P.-F. Li, Circular RNA mediates cardiomyocyte death via miRNA-dependent
REFERENCES AND NOTES upregulation of MTP18 expression. Cell Death Differ. 24, 1111–1120 (2017).
1. R. Lozano, M. Naghavi, K. Foreman, S. Lim, K. Shibuya, V. Aboyans, J. Abraham, T. Adair,
18. K. Wang, C.-Y. Liu, X.-J. Zhang, C. Feng, L.-Y. Zhou, Y. Zhao, P.-F. Li, MiR-361-regulated
R. Aggarwal, S. Y. Ahn, M. Alvarado, H. R. Anderson, L. M. Anderson, K. G. Andrews,
prohibitin inhibits mitochondrial fission and apoptosis and protects heart from ischemia
C. Atkinson, L. M. Baddour, S. Barker-Collo, D. H. Bartels, M. L. Bell, E. J. Benjamin,
injury. Cell Death Differ. 22, 1058–1068 (2015).
D. Bennett, K. Bhalla, B. Bikbov, A. B. Abdulhak, G. Birbeck, F. Blyth, I. Bolliger, S. Boufous,
19. K. Wang, B. Long, L.-Y. Zhou, F. Liu, Q.-Y. Zhou, C.-Y. Liu, Y.-Y. Fan, P.-F. Li, CARL lncRNA
C. Bucello, M. Burch, P. Burney, J. Carapetis, H. Chen, D. Chou, S. S. Chugh, L. E. Coffeng,
inhibits anoxia-induced mitochondrial fission and apoptosis in cardiomyocytes by
S. D. Colan, S. Colquhoun, K. E. Colson, J. Condon, M. D. Connor, L. T. Cooper, M. Corriere,
impairing miR-539-dependent PHB2 downregulation. Nat. Commun. 5, 3596 (2014).
M. Cortinovis, K. C. de Vaccaro, W. Couser, B. C. Cowie, M. H. Criqui, M. Cross,
20. K. Wang, L.-Y. Zhou, J.-X. Wang, Y. Wang, T. Sun, B. Zhao, Y.-J. Yang, T. An, B. Long, N. Li,
K. C. Dabhadkar, N. Dahodwala, D. De Leo, L. Degenhardt, A. Delossantos, J. Denenberg,
C.-Y. Liu, Y. Gong, J.-N. Gao, Y.-H. Dong, J. Zhang, P.-F. Li, E2F1-dependent MIR-421
D. C. D. Jarlais, S. D. Dharmaratne, E. R. Dorsey, T. Driscoll, H. Duber, B. Ebel, P. J. Erwin,
regulates mitochondrial fragmentation and myocardial infarction by targeting Pink1.
P. Espindola, M. Ezzati, V. Feigin, A. D. Flaxman, M. H. Forouzanfar, F. G. R. Fowkes,
Nat. Commun. 6, 7619 (2015).
R. Franklin, M. Fransen, M. K. Freeman, S. E. Gabriel, E. Gakidou, F. Gaspari, R. F. Gillum,
21. P. Hang, C. Sun, J. Guo, J. Zhao, Z. Du, BDNF-mediates down-regulation of microRNA-195
D. Gonzalez-Medina, Y. A. Halasa, D. Haring, J. E. Harrison, R. Havmoeller, R. J. Hay,
inhibits ischemic cardiac apoptosis in rats. Int. J. Biol. Sci. 12, 979–989 (2016).
B. Hoen, P. J. Hotez, D. Hoy, K. H. Jacobsen, S. L. James, R. Jasrasaria, S. Jayaraman,
N. Johns, G. Karthikeyan, N. Kassebaum, A. Keren, J.-P. Khoo, L. M. Knowlton, 22. Y. Tang, J. Zheng, Y. Sun, Z. Wu, Z. Liu, G. Huang, MicroRNA-1 regulates cardiomyocyte
O. Kobusingye, A. Koranteng, R. Krishnamurthi, M. Lipnick, S. E. Lipshultz, S. L. Ohno, apoptosis by targeting Bcl-2. Int. Heart J. 50, 377–387 (2009).
J. Mabweijano, M. F. MacIntyre, L. Mallinger, L. March, G. B. Marks, R. Marks, A. Matsumori, 23. W. Yang, Y. Han, C. Yang, Y. Chen, W. Zhao, X. Su, K. Yang, W. Jin, MicroRNA-19b-1
R. Matzopoulos, B. M. Mayosi, J. H. McAnulty, M. M. McDermott, J. M. Grath, G. A. Mensah, reverses ischaemia-induced heart failure by inhibiting cardiomyocyte apoptosis
T. R. Merriman, C. Michaud, M. Miller, T. R. Miller, C. Mock, A. O. Mocumbi, A. A. Mokdad, and targeting Bcl2 l11/BIM. Heart Vessels 34, 1221–1229 (2019).
A. Moran, K. Mulholland, M. N. Nair, L. Naldi, K. M. V. Narayan, K. Nasseri, P. Norman, 24. L. Qian, L. W. Van Laake, Y. Huang, S. Liu, M. F. Wendland, D. Srivastava, miR-24 inhibits
M. O'Donnell, S. B. Omer, K. Ortblad, R. Osborne, D. Ozgediz, B. Pahari, J. D. Pandian, apoptosis and represses Bim in mouse cardiomyocytes. J. Exp. Med. 208, 549–560 (2011).
A. P. Rivero, R. P. Padilla, F. Perez-Ruiz, N. Perico, D. Phillips, K. Pierce, C. Arden Pope III, 25. A. S. Bayoumi, K.-M. Park, Y. Wang, J.-P. Teoh, T. Aonuma, Y. Tang, H. Su, N. L. Weintraub,
E. Porrini, F. Pourmalek, M. Raju, D. Ranganathan, J. T. Rehm, D. B. Rein, G. Remuzzi, I.-M. Kim, A carvedilol-responsive microRNA, miR-125b-5p protects the heart from acute
F. P. Rivara, T. Roberts, F. R. De León, L. C. Rosenfeld, L. Rushton, R. L. Sacco, J. A. Salomon, myocardial infarction by repressing pro-apoptotic bak1 and klf13 in cardiomyocytes.
U. Sampson, E. Sanman, D. C. Schwebel, M. Segui-Gomez, D. S. Shepard, D. Singh, J. Mol. Cell. Cardiol. 114, 72–82 (2018).
J. Singleton, K. Sliwa, E. Smith, A. Steer, J. A. Taylor, B. Thomas, I. M. Tleyjeh, J. A. Towbin, 26. S. Song, H.-H. Seo, S.-Y. Lee, C. Y. Lee, J. Lee, K.-J. Yoo, C. Yoon, E. Choi, K.-C. Hwang, S. Lee,
T. Truelsen, E. A. Undurraga, N. Venketasubramanian, L. Vijayakumar, T. Vos, G. R. Wagner, MicroRNA-17-mediated down-regulation of apoptotic protease activating factor 1
M. Wang, W. Wang, K. Watt, M. A. Weinstock, R. Weintraub, J. D. Wilkinson, A. D. Woolf, attenuates apoptosome formation and subsequent apoptosis of cardiomyocytes.
S. Wulf, P.-H. Yeh, P. Yip, A. Zabetian, Z.-J. Zheng, A. D. Lopez, C. J. L. Murray, Biochem. Biophys. Res. Commun. 465, 299–304 (2015).
M. A. Al Mazroa, Z. A. Memish, Global and regional mortality from 235 causes of death 27. Y. Li, S. Ren, J. Xia, Y. Wei, Y. Xi, EIF4A3-induced circ-BNIP3 aggravated hypoxia-induced
for 20 age groups in 1990 and 2010: A systematic analysis for the Global Burden injury of H9c2 cells by targeting miR-27a-3p/BNIP3. Mol. Ther. Nucleic Acids 19, 533–545
of Disease Study 2010. Lancet 380, 2095–2128 (2012). (2020).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 14 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

28. S.-F. He, H.-J. Zhu, Z.-Y. Han, H. Wu, S.-Y. Jin, M. G. Irwin, Y. Zhang, MicroRNA-133b-5p is 52. S. Shin, J.-W. Choi, H. Moon, C. Y. Lee, J.-H. Park, J. Lee, H.-H. Seo, G. Han, S. Lim, S. Lee,
involved in cardioprotection of morphine preconditioning in rat cardiomyocytes by S. W. Kim, K.-C. Hwang, Simultaneous suppression of multiple programmed cell death
targeting fas. Can. J. Cardiol. 32, 996–1007 (2016). pathways by miRNA-105 in cardiac ischemic injury. Mol. Ther. Nucleic Acids 14, 438–449
29. Y. Wang, M.-M. Tian, C.-J. Mi, K.-L. Chen, Y.-C. Ji, L. Wang, J. Zhang, K. Cheng, Exercise (2019).
protects the heart against myocardial infarction through upregulation of miR-1192. 53. K. Wang, F. Liu, C.-Y. Liu, T. An, J. Zhang, L.-Y. Zhou, M. Wang, Y.-H. Dong, N. Li, J.-N. Gao,
Biochem. Biophys. Res. Commun. 521, 1061–1069 (2019). Y.-F. Zhao, P.-F. Li, The long noncoding RNA NRF regulates programmed necrosis
30. Q. Li, J. Yang, J. Zhang, X.-W. Liu, C.-J. Yang, Z.-X. Fan, H.-B. Wang, Y. Yang, T. Zheng, and myocardial injury during ischemia and reperfusion by targeting miR-873. Cell Death
J. Yang, Inhibition of microRNA-327 ameliorates ischemia/reperfusion injury-induced Differ. 23, 1394–1405 (2016).
cardiomyocytes apoptosis through targeting apoptosis repressor with caspase 54. D. Qin, X. Wang, Y. Li, L. Yang, R. Wang, J. Peng, K. Essandoh, X. Mu, T. Peng, Q. Han,
recruitment domain. J. Cell. Physiol. 235, 3753–3767 (2020). K.-J. Yu, G.-C. Fan, MicroRNA-223-5p and -3p cooperatively suppress necroptosis
31. T. F. Franke, C. P. Hornik, L. Segev, G. A. Shostak, C. Sugimoto, PI3K/Akt and apoptosis: in ischemic/reperfused hearts. J. Biol. Chem. 291, 20247–20259 (2016).
Size matters. Oncogene 22, 8983–8998 (2003). 55. J.-X. Wang, X.-J. Zhang, Q. Li, K. Wang, Y. Wang, J.-Q. Jiao, C. Feng, S. Teng, L.-Y. Zhou,
32. L. Zhao, X. R. Yang, X. Han, MicroRNA-146b induces the Pi3k/Akt/NF-B signaling Y. Gong, Z.-X. Zhou, J. Liu, J.-L. Wang, P.-f. Li, MicroRNA-103/107 regulate programmed
pathway to reduce vascular inflammation and apoptosis in myocardial infarction by necrosis and myocardial ischemia/reperfusion injury through targeting FADD. Circ. Res.
targeting PTEN. Exp. Ther. Med. 17, 1171–1181 (2019). 117, 352–363 (2015).
33. L. Yao, Q. Zhou, L. Wang, G. Hou, MicroRNA-182-5p protects H9c2 cardiomyocytes 56. K. Wang, F. Liu, L.-Y. Zhou, S.-L. Ding, B. Long, C.-Y. Liu, T. Sun, Y.-Y. Fan, L. Sun, P.-F. Li,
from hypoxia-induced apoptosis by down-regulation of PTEN. Int. J. Clin. Exp. Pathol. 10, MiR-874 regulates myocardial necrosis by targeting caspase-8. Cell Death Dis. 4, e709 (2013).

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


5220–5226 (2017). 57. K. Wang, C.-Y. Liu, L.-Y. Zhou, J.-X. Wang, M. Wang, B. Zhao, W.-K. Zhao, S.-J. Xu, L.-H. Fan,
34. X. Xing, S. Guo, G. Zhang, Y. Liu, S. Bi, X. Wang, Q. Lu, miR-26a-5p protects against X.-J. Zhang, C. Feng, C.-Q. Wang, Y.-F. Zhao, P.-F. Li, APF lncRNA regulates autophagy
myocardial ischemia/reperfusion injury by regulating the PTEN/PI3K/AKT signaling and myocardial infarction by targeting miR-188-3p. Nat. Commun. 6, 6779 (2015).
pathway. Braz. J. Med. Biol. Res. 53, e9106 (2020). 58. J. Liu, M. Jiang, S. Deng, J. Lu, H. Huang, Y. Zhang, P. Gong, X. Shen, H. Ruan, M. Jin,
35. Z.-P. Ke, P. Xu, Y. Shi, A.-M. Gao, MicroRNA-93 inhibits ischemia-reperfusion induced H. Wang, miR-93-5p-containing exosomes treatment attenuates acute myocardial
cardiomyocyte apoptosis by targeting PTEN. Oncotarget 7, 28796–28805 (2016). infarction-induced myocardial damage. Mol. Ther. Nucleic Acids 11, 103–115 (2018).
36. C.-L. Song, B. Liu, H.-Y. Diao, Y.-F. Shi, J.-C. Zhang, Y.-X. Li, N. Liu, Y.-P. Yu, G. Wang, 59. S. K. Gupta, A. Foinquinos, S. Thum, J. Remke, K. Zimmer, C. Bauters, P. de Groote,
J.-P. Wang, Q. Li, Down-regulation of microRNA-320 suppresses cardiomyocyte apoptosis R. A. Boon, L. J. de Windt, S. Preissl, L. Hein, S. Batkai, F. Pinet, T. Thum, Preclinical
and protects against myocardial ischemia and reperfusion injury by targeting IGF-1. development of a microRNA-based therapy for elderly patients with myocardial
Oncotarget 7, 39740–39757 (2016). infarction. J. Am. Coll. Cardiol. 68, 1557–1571 (2016).
37. R. Sun, L. Zhang, Long non-coding RNA MALAT1 regulates cardiomyocytes apoptosis 60. A. Ucar, S. K. Gupta, J. Fiedler, E. Erikci, M. Kardasinski, S. Batkai, S. Dangwal,
after hypoxia/reperfusion injury via modulating miR-200a-3p/PDCD4 axis. Biomed. R. Kumarswamy, C. Bang, A. Holzmann, J. Remke, M. Caprio, C. Jentzsch, S. Engelhardt,
Pharmacother. 111, 1036–1045 (2019). S. Geisendorf, C. Glas, T. G. Hofmann, M. Nessling, K. Richter, M. Schiffer, L. Carrier,
38. K. M. Luther, L. Haar, M. M. Guinness, Y. Wang, T. L. Lynch IV, A. Phan, Y. Song, Z. Shen, L. C. Napp, J. Bauersachs, K. Chowdhury, T. Thum, The miRNA-212/132 family regulates
G. Gardner, G. Kuffel, X. Ren, M. J. Zilliox, W. K. Jones, Exosomal miR-21a-5p mediates both cardiac hypertrophy and cardiomyocyte autophagy. Nat. Commun. 3, 1078 (2012).
cardioprotection by mesenchymal stem cells. J. Mol. Cell. Cardiol. 119, 125–137 (2018). 61. Q. Su, Y. Liu, X.-W. Lv, Z.-L. Ye, Y.-H. Sun, B.-H. Kong, Z.-B. Qin, Inhibition of lncRNA TUG1
39. H. Gu, Z. Liu, Y. Li, Y. Xie, J. Yao, Y. Zhu, J. Xu, Q. Dai, C. Zhong, H. Zhu, S. Ding, L. Zhou, upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion
Serum-derived extracellular vesicles protect against acute myocardial infarction by via targeting HMGB1- and Rac1-induced autophagy. J. Mol. Cell. Cardiol. 133, 12–25
regulating miR-21/PDCD4 signaling pathway. Front. Physiol. 9, 348 (2018). (2019).
40. Y. Cheng, X. Liu, S. Zhang, Y. Lin, J. Yang, C. Zhang, MicroRNA-21 protects against 62. S. Hu, S. Cao, Z. Tong, J. Liu, FGF21 protects myocardial ischemia-reperfusion injury
the H2O2-induced injury on cardiac myocytes via its target gene PDCD4. J. Mol. Cell. through reduction of miR-145-mediated autophagy. Am. J. Transl. Res. 10, 3677–3688 (2018).
Cardiol. 47, 5–14 (2009). 63. Q. Chen, Y. Zhou, A. M. Richards, P. Wang, Up-regulation of miRNA-221 inhibits hypoxia/
41. J. Wang, Z. Jia, C. Zhang, M. Sun, W. Wang, P. Chen, K. Ma, Y. Zhang, X. Li, C. Zhou, reoxygenation-induced autophagy through the DDIT4/mTORC1 and Tp53inp1/p62
miR-499 protects cardiomyocytes from H2O2-induced apoptosis via its effects on Pdcd4 pathways. Biochem. Biophys. Res. Commun. 474, 168–174 (2016).
and Pacs2. RNA Biol. 11, 339–350 (2014). 64. Y. Li, R. Yang, B. Guo, H. Zhang, H. Zhang, S. Liu, Y. Li, Exosomal miR-301 derived
42. J. Ma, J. Zhang, Y. Wang, K. Long, X. Wang, L. Jin, Q. Tang, L. Zhu, G. Tang, X. Li, M. Li, from mesenchymal stem cells protects myocardial infarction by inhibiting myocardial
MiR-532-5p alleviates hypoxia-induced cardiomyocyte apoptosis by targeting PDCD4. autophagy. Biochem. Biophys. Res. Commun. 514, 323–328 (2019).
Gene 675, 36–43 (2018). 65. B. Lin, D. Feng, J. Xu, Cardioprotective effects of microRNA-18a on acute myocardial
43. Z. Pan, X. Sun, J. Ren, X. Li, X. Gao, C. Lu, Y. Zhang, H. Sun, Y. Wang, H. Wang, J. Wang, infarction by promoting cardiomyocyte autophagy and suppressing cellular senescence
L. Xie, Y. Lu, B. Yang, miR-1 exacerbates cardiac ischemia-reperfusion injury in mouse via brain derived neurotrophic factor. Cell Biosci. 9, 38 (2019).
models. PLOS ONE 7, e50515 (2012). 66. Z. Li, Y. Song, L. Liu, N. Hou, X. An, D. Zhan, Y. Li, L. Zhou, P. Li, L. Yu, J. Xia, Y. Zhang,
44. H. Hong, T. Tao, S. Chen, C. Liang, Y. Qiu, Y. Zhou, R. Zhang, MicroRNA-143 promotes J. Wang, X. Yang, MiR-199a impairs autophagy and induces cardiac hypertrophy through
cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase mTOR activation. Cell Death Differ. 24, 1205–1213 (2017).
Cepsilon. Basic Res. Cardiol. 112, 60 (2017). 67. Y. Yang, Y. Li, X. Chen, X. Cheng, Y. Liao, X. Yu, Exosomal transfer of miR-30a between
45. Y. Wang, M. Men, W. Yang, H. Zheng, S. Xue, MiR-31 downregulation protects against cardiomyocytes regulates autophagy after hypoxia. J. Mol. Med. 94, 711–724 (2016).
cardiac ischemia/reperfusion injury by targeting protein kinase C epsilon (PKC) directly. 68. X. Guo, X. Wu, Y. Han, E. Tian, J. Cheng, LncRNA MALAT1 protects cardiomyocytes
Cell. Physiol. Biochem. 36, 179–190 (2015). from isoproterenol-induced apoptosis through sponging miR-558 to enhance
46. X. Meng, Y. Ji, Z. Wan, B. Zhao, C. Feng, J. Zhao, H. Li, Y. Song, Inhibition of miR-363 ULK1-mediated protective autophagy. J. Cell. Physiol. 234, 10842–10854 (2019).
protects cardiomyocytes against hypoxia-induced apoptosis through regulation 69. Y. Tian, Y. Liu, T. Wang, N. Zhou, J. Kong, L. Chen, M. Snitow, M. Morley, D. Li, N. Petrenko,
of Notch signaling. Biomed. Pharmacother. 90, 509–516 (2017). S. Zhou, M. Lu, E. Gao, W. J. Koch, K. M. Stewart, E. E. Morrisey, A microRNA-Hippo
47. H. Xu, L. Jin, Y. Chen, J. Li, Downregulation of microRNA-429 protects cardiomyocytes pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice.
against hypoxia-induced apoptosis by increasing Notch1 expression. Int. J. Mol. Med. 37, Sci. Transl. Med. 7, 279ra38 (2015).
1677–1685 (2016). 70. A. Eulalio, M. Mano, M. D. Ferro, L. Zentilin, G. Sinagra, S. Zacchigna, M. Giacca,
48. Z. Chen, X. Su, Y. Shen, Y. Jin, T. Luo, I.-M. Kim, N. L. Weintraub, Y. Tang, MiR322 mediates Functional screening identifies miRNAs inducing cardiac regeneration. Nature 492,
cardioprotection against ischemia/reperfusion injury via FBXW7/notch pathway. J. Mol. 376–381 (2012).
Cell. Cardiol. 133, 67–74 (2019). 71. C. Torrini, R. J. Cubero, E. Dirkx, L. Braga, H. Ali, G. Prosdocimo, M. I. Gutierrez, C. Collesi,
49. K. Yan, T. An, M. Zhai, Y. Huang, Q. Wang, Y. Wang, R. Zhang, T. Wang, J. Liu, Y. Zhang, D. Licastro, L. Zentilin, M. Mano, S. Zacchigna, M. Vendruscolo, M. Marsili, A. Samal,
J. Zhang, K. Wang, Mitochondrial miR-762 regulates apoptosis and myocardial infarction M. Giacca, Common regulatory pathways mediate activity of microRNAs inducing
by impairing ND2. Cell Death Dis. 10, 500 (2019). cardiomyocyte proliferation. Cell Rep. 27, 2759–2771.e5 (2019).
50. D. Lin, B. Cui, J. Ma, J. Ren, MiR-183-5p protects rat hearts against myocardial ischemia/ 72. K. Gabisonia, G. Prosdocimo, G. D. Aquaro, L. Carlucci, L. Zentilin, I. Secco, H. Ali, L. Braga,
reperfusion injury through targeting VDAC1. Biofactors 46, 83–93 (2020). N. Gorgodze, F. Bernini, S. Burchielli, C. Collesi, L. Zandonà, G. Sinagra, M. Piacenti,
51. Q. Su, Y. Liu, X.-W. Lv, R.-X. Dai, X.-H. Yang, B.-H. Kong, LncRNA TUG1 mediates ischemic S. Zacchigna, R. Bussani, F. A. Recchia, M. Giacca, MicroRNA therapy stimulates
myocardial injury by targeting miR-132-3p/HDAC3 axis. Am. J. Physiol. Circ. Physiol. 318, uncontrolled cardiac repair after myocardial infarction in pigs. Nature 569, 418–422
H332–H344 (2020). (2019).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 15 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

73. Y. Xiao, Y. Zhang, Y. Chen, J. Li, Z. Zhang, Y. Sun, H. Shen, Z. Zhao, Z. Huang, W. Zhang, phenotype and reversibility of fibrosis in patients with end-stage heart failure. J. Am. Coll.
W. Chen, Z. Shen, Inhibition of microRNA-9-5p protects against cardiac remodeling Cardiol. 73, 2267–2282 (2019).
following myocardial infarction in mice. Hum. Gene Ther. 30, 286–301 (2019). 96. Z. Pan, X. Sun, H. Shan, N. Wang, J. Wang, J. Ren, S. Feng, L. Xie, C. Lu, Y. Yuan,
74. Y. Yang, H.-W. Cheng, Y. Qiu, D. Dupee, M. Noonan, Y.-D. Lin, S. Fisch, K. Unno, K.-I. Sereti, Y. Zhang, Y. Wang, Y. Lu, B. Yang, miR-101 inhibited post-infarct cardiac fibrosis
R. Liao, MicroRNA-34a plays a key role in cardiac repair and regeneration following and improved left ventricular compliance via FOS/TGF1 pathway. Circulation 126,
myocardial infarction. Circ. Res. 117, 450–459 (2015). 840–850 (2012).
75. Y. Hu, G. Jin, B. Li, Y. Chen, L. Zhong, G. Chen, X. Chen, J. Zhong, W. Liao, Y. Liao, Y. Wang, 97. X. Zhao, K. Wang, Y. Liao, Q. Zeng, Y. Li, F. Hu, Y. Liu, K. Meng, C. Qian, Q. Zhang, H. Guan,
J. Bin, Suppression of miRNA let-7i-5p promotes cardiomyocyte proliferation and repairs K. Feng, Y. Zhou, Y. Du, Z. Chen, MicroRNA-101a inhibits cardiac fibrosis induced by
heart function post injury by targetting CCND2 and E2F2. Clin. Sci. 133, 425–441 (2019). hypoxia via targeting TGF RI on cardiac fibroblasts. Cell. Physiol. Biochem. 35, 213–226 (2015).
76. A. Borden, J. Kurian, E. Nickoloff, Y. Yang, C. D. Troupes, J. Ibetti, A. M. Lucchese, E. Gao, 98. L. Tao, Y. Bei, P. Chen, Z. Lei, S. Fu, H. Zhang, J. Xu, L. Che, X. Chen, J. P. G. Sluijter, S. Das,
S. Mohsin, W. J. Koch, S. R. Houser, R. Kishore, M. Khan, Transient introduction of miR-294 D. Cretoiu, B. Xu, J. Zhong, J. Xiao, X. Li, Crucial role of miR-433 in regulating cardiac
in the heart promotes cardiomyocyte cell cycle reentry after injury. Circ. Res. 125, 14–25 fibrosis. Theranostics 6, 2068–2083 (2016).
(2019). 99. J. Wang, W. Huang, R. Xu, Y. Nie, X. Cao, J. Meng, X. Xu, S. Hu, Z. Zheng, MicroRNA-24
77. W. Huang, Y. Feng, J. Liang, H. Yu, C. Wang, B. Wang, M. Wang, L. Jiang, W. Meng, W. Cai, regulates cardiac fibrosis after myocardial infarction. J. Cell. Mol. Med. 16, 2150–2160 (2012).
M. Medvedovic, J. Chen, C. Paul, W. S. Davidson, S. Sadayappan, P. J. Stambrook, X.-Y. Yu, 100. Y. Hong, H. Cao, Q. Wang, J. Ye, L. Sui, J. Feng, X. Cai, H. Song, X. Zhang, X. Chen, MiR-22
Y. Wang, Loss of microRNA-128 promotes cardiomyocyte proliferation and heart may suppress fibrogenesis by targeting TGFR I in cardiac fibroblasts. Cell. Physiol.
regeneration. Nat. Commun. 9, 700 (2018). Biochem. 40, 1345–1353 (2016).

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


78. J. Zhang, L. Wu, Z. Li, G. Fu, miR-1231 exacerbates arrhythmia by targeting 101. H. Liang, C. Zhang, T. Ban, Y. Liu, L. Mei, X. Piao, D. Zhao, Y. Lu, W. Chu, B. Yang, A novel
calciumchannel gene CACNA2D2 in myocardial infarction. Am. J. Transl. Res. 9, 1822–1833 reciprocal loop between microRNA-21 and TGFRIII is involved in cardiac fibrosis.
(2017). Int. J. Biochem. Cell Biol. 44, 2152–2160 (2012).
79. X. Liu, Y. Zhang, W. Du, H. Liang, H. He, L. Zhang, Z. Pan, X. Li, C. Xu, Y. Zhou, L. Wang, 102. W. Du, H. Liang, X. Gao, X. Li, Y. Zhang, Z. Pan, C. Li, Y. Wang, Y. Liu, W. Yuan, N. Ma,
M. Qian, T. Liu, H. Yin, Y. Lu, B. Yang, H. Shan, MiR-223-3p as a novel microRNA regulator W. Chu, H. Shan, Y. Lu, MicroRNA-328, a potential anti-fibrotic target in cardiac interstitial
of expression of voltage-gated K+ Channel Kv4.2 in acute myocardial infarction. Cell. fibrosis. Cell. Physiol. Biochem. 39, 827–836 (2016).
Physiol. Biochem. 39, 102–114 (2016). 103. J. Yuan, H. Chen, D. Ge, Y. Xu, H. Xu, Y. Yang, M. Gu, Y. Zhou, J. Zhu, T. Ge, Q. Chen, Y. Gao,
80. X. Zhou, F. Sun, S. Luo, W. Zhao, T. Yang, G. Zhang, M. Gao, R. Lu, Y. Shu, W. Mu, Y. Wang, X. Li, Y. Zhao, Mir-21 promotes cardiac fibrosis after myocardial infarction via
Y. Zhuang, F. Ding, C. Xu, Y. Lu, Let-7a is an antihypertrophic regulator in the heart via targeting Smad7. Cell. Physiol. Biochem. 42, 2207–2219 (2017).
targeting calmodulin. Int. J. Biol. Sci. 13, 22–31 (2017). 104. Y. Huang, Y. Qi, J.-Q. Du, D.-f. Zhang, MicroRNA-34a regulates cardiac fibrosis after
81. Y. Yang, D. P. Del Re, N. Nakano, S. Sciarretta, P. Zhai, J. Park, D. Sayed, A. Shirakabe, myocardial infarction by targeting Smad4. Expert Opin. Ther. Targets 18, 1355–1365
S. Matsushima, Y. Park, B. Tian, M. Abdellatif, J. Sadoshima, MIR-206 mediates (2014).
YAP-induced cardiac hypertrophy and survival. Circ. Res. 117, 891–904 (2015). 105. X. Wang, M. B. Morelli, A. Matarese, C. Sardu, G. Santulli, Cardiomyocyte-derived
82. J. Yang, F. Hu, X. Fu, Z. Jiang, W. Zhang, K. Chen, MiR-128/SOX7 alleviates myocardial exosomal microRNA-92a mediates post-ischemic myofibroblast activation both in vitro
ischemia injury by regulating IL-33/sST2 in acute myocardial infarction. Biol. Chem. 400, and ex vivo. ESC Hear. Fail. 7, 285–289 (2020).
533–544 (2019). 106. M. B. Morelli, J. Shu, C. Sardu, A. Matarese, G. Santulli, Cardiosomal microRNAs are
83. A. Li, Y. Yu, X. Ding, Y. Qin, Y. Jiang, X. Wang, G. Liu, X. Chen, E. Yue, X. Sun, S. M. Zahra, essential in post-infarction myofibroblast phenoconversion. Int. J. Mol. Sci. 21, 201 (2020).
Y. Yan, L. Ren, S. Wang, L. Chai, Y. Bai, B. Yang, MiR-135b protects cardiomyocytes 107. L. E. Dorn, J. M. Petrosino, P. Wright, F. Accornero, CTGF/CCN2 is an autocrine regulator
from infarction through restraining the NLRP3/caspase-1/IL-1 pathway. Int. J. Cardiol. of cardiac fibrosis. J. Mol. Cell. Cardiol. 121, 205–211 (2018).
307, 137–145 (2019). 108. K. E. Lipson, C. Wong, Y. Teng, S. Spong, CTGF is a central mediator of tissue remodeling
84. M. Yuan, L. Zhang, F. You, J. Zhou, Y. Ma, F. Yang, L. Tao, MiR-145-5p regulates and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenesis Tissue Repair
hypoxia-induced inflammatory response and apoptosis in cardiomyocytes by targeting 5, S24 (2012).
CD40. Mol. Cell. Biochem. 431, 123–131 (2017). 109. R. F. Duisters, A. J. Tijsen, B. Schroen, J. J. Leenders, V. Lentink, I. van der Made, V. Herias,
85. N. A. Turner, A. Das, P. Warburton, D. J. O’Regan, S. G. Ball, K. E. Porter, Interleukin-1 R. E. van Leeuwen, M. W. Schellings, P. Barenbrug, J. G. Maessen, S. Heymans, Y. M. Pinto,
stimulates proinflammatory cytokine expression in human cardiac myofibroblasts. E. E. Creemers, miR-133 and miR-30 regulate connective tissue growth factor:
Am. J. Physiol. Hear. Circ. Physiol. 297, H1117–H1127 (2009). Implications for a role of microRNAs in myocardial matrix remodeling. Circ. Res. 104,
86. M. Kawaguchi, M. Takahashi, T. Hata, Y. Kashima, F. Usui, H. Morimoto, A. Izawa, 170–178 (2009).
Y. Takahashi, J. Masumoto, J. Koyama, M. Hongo, T. Noda, J. Nakayama, J. Sagara, 110. L. Chen, Q. Ji, H. Zhu, Y. Ren, Z. Fan, N. Tian, miR-30a attenuates cardiac fibrosis in rats
S. Taniguchi, U. Ikeda, Inflammasome activation of cardiac fibroblasts is essential with myocardial infarction by inhibiting CTGF. Exp. Ther. Med. 15, 4318–4324 (2018).
for myocardial ischemia/reperfusion injury. Circulation 123, 594–604 (2011). 111. T. Thum, C. Gross, J. Fiedler, T. Fischer, S. Kissler, M. Bussen, P. Galuppo, S. Just,
87. M. L. Lindsey, R. Zamilpa, Temporal and spatial expression of matrix metalloproteinases W. Rottbauer, S. Frantz, M. Castoldi, J. Soutschek, V. Koteliansky, A. Rosenwald,
and tissue inhibitors of metalloproteinases following myocardial infarction. Cardiovasc. M. A. Basson, J. D. Licht, J. T. R. Pena, S. H. Rouhanifard, M. U. Muckenthaler, T. Tuschl,
Ther. 30, 31–41 (2012). G. R. Martin, J. Bauersachs, S. Engelhardt, MicroRNA-21 contributes to myocardial disease
88. P. J. Lijnen, V. V. Petrov, R. H. Fagard, Induction of cardiac fibrosis by transforming growth by stimulating MAP kinase signalling in fibroblasts. Nature 456, 980–984 (2008).
factor-1. Mol. Genet. Metab. 71, 418–435 (2000). 112. O. Adam, B. Löhfelm, T. Thum, S. K. Gupta, S. L. Puhl, H. J. Schäfers, M. Böhm, U. Laufs, Role
89. Y. Sun, J. Q. Zhang, J. Zhang, F. J. A. Ramires, Angiotensin II, transforming growth of miR-21 in the pathogenesis of atrial fibrosis. Basic Res. Cardiol. 107, 278 (2012).
factor-1 and repair in the infarcted heart. J. Mol. Cell. Cardiol. 30, 1559–1569 (1998). 113. S. Cardin, E. Guasch, X. Luo, P. Naud, K. L. Quang, Y. F. Shi, J.-C. Tardif, P. Comtois, S. Nattel,
90. H. Ye, P.-C. Cai, Q. Zhou, W.-L. Ma, Transforming growth factor-1 suppresses Role for MicroRNA-21 in atrial profibrillatory fibrotic remodeling associated
the up-regulation of matrix metalloproteinase-2 by lung fibroblasts in response to tumor with experimental postinfarction heart failure. Circ. Arrhythm. Electrophysiol. 5,
necrosis factor-. Wound Repair Regen. 19, 392–399 (2011). 1027–1035 (2012).
91. A.-H. Li, P. P. Liu, F. J. Villarreal, R. A. Garcia, Dynamic changes in myocardial matrix 114. X. Yuan, J. Pan, L. Wen, B. Gong, J. Li, H. Gao, W. Tan, S. Liang, H. Zhang, X. Wang,
and relevance to disease: Translational perspectives. Circ. Res. 114, 916–927 (2014). MiR-144-3p enhances cardiac fibrosis after myocardial infarction by targeting PTEN.
92. C. K. Nagaraju, E. Dries, N. Popovic, A. A. Singh, P. Haemers, H. L. Roderick, P. Claus, Front. Cell Dev. Biol. 7, 249 (2019).
K. R. Sipido, R. B. Driesen, Global fibroblast activation throughout the left ventricle but 115. T. Boettger, N. Beetz, S. Kostin, J. Schneider, M. Krüger, L. Hein, T. Braun, Acquisition
localized fibrosis after myocardial infarction. Sci. Rep. 7, 10801 (2017). of the contractile phenotype by murine arterial smooth muscle cells depends
93. N. G. Frangogiannis, L. H. Michael, M. L. Entman, Myofibroblasts in reperfused myocardial on the Mir143/145 gene cluster. J. Clin. Invest. 119, 2634–2647 (2009).
infarcts express the embryonic form of smooth muscle myosin heavy chain (SMemb). 116. C. Li, J. Li, K. Xue, J. Zhang, C. Wang, Q. Zhang, X. Chen, C. Gao, X. Yu, L. Sun, MicroRNA-
Cardiovasc. Res. 48, 89–100 (2000). 143-3p promotes human cardiac fibrosis via targeting sprouty3 after myocardial
94. X. Fu, H. Khalil, O. Kanisicak, J. G. Boyer, R. J. Vagnozzi, B. D. Maliken, M. A. Sargent, infarction. J. Mol. Cell. Cardiol. 129, 281–292 (2019).
V. Prasad, I. Valiente-Alandi, B. C. Blaxall, J. D. Molkentin, Specialized fibroblast 117. Y.-S. Wang, S.-H. Li, J. Guo, A. Mihic, J. Wu, L. Sun, K. Davis, R. D. Weisel, R.-K. Li, Role
differentiated states underlie scar formation in the infarcted mouse heart. J. Clin. Invest. of miR-145 in cardiac myofibroblast differentiation. J. Mol. Cell. Cardiol. 66, 94–105 (2014).
128, 2127–2143 (2018). 118. N. Liu, S. Bezprozvannaya, A. H. Williams, X. Qi, J. A. Richardson, R. Bassel-Duby,
95. C. K. Nagaraju, E. L. Robinson, M. Abdesselem, S. Trenson, E. Dries, G. Gilbert, S. Janssens, E. N. Olson, microRNA-133a regulates cardiomyocyte proliferation and suppresses
J. Van Cleemput, F. Rega, B. Meyns, H. L. Roderick, R. B. Driesen, K. R. Sipido, Myofibroblast smooth muscle gene expression in the heart. Genes Dev. 22, 3242–3254 (2008).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 16 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

119. E. Boštjančič, N. Zidar, D. Štajer, D. Glavač, MicroRNAs miR-1, miR-133a, miR-133b and 141. M. Meloni, M. Marchetti, K. Garner, B. Littlejohns, G. Sala-Newby, N. Xenophontos, I. Floris,
miR-208 are dysregulated in human myocardial infarction. Cardiology 115, 163–169 (2010). M.-S. Suleiman, P. Madeddu, A. Caporali, C. Emanueli, Local inhibition of microRNA-24
120. E. van Rooij, L. B. Sutherland, J. E. Thatcher, J. M. DiMaio, R. H. Naseem, W. S. Marshall, improves reparative angiogenesis and left ventricle remodeling and function in mice
J. A. Hill, E. N. Olson, Dysregulation of microRNAs after myocardial infarction reveals a role with myocardial infarction. Mol. Ther. 21, 1390–1402 (2013).
of miR-29 in cardiac fibrosis. Proc. Natl. Acad. Sci. U.S.A. 105, 13027–13032 (2008). 142. S. Liu, J. Chen, J. Shi, W. Zhou, L. Wang, W. Fang, Y. Zhong, X. Chen, Y. Chen, A. Sabri,
121. J. D. Port, L. A. Walker, J. Polk, K. Nunley, P. M. Buttrick, C. C. Sucharov, Temporal S. Liu, M1-like macrophage-derived exosomes suppress angiogenesis and exacerbate
expression of miRNAs and mRNAs in a mouse model of myocardial infarction. Physiol. cardiac dysfunction in a myocardial infarction microenvironment. Basic Res. Cardiol. 115,
Genomics 43, 1087–1095 (2011). 22 (2020).
122. G. Castoldi, C. R. T. Di Gioia, C. Bombardi, D. Catalucci, B. Corradi, M. G. Gualazzi, 143. V. Joris, E. L. Gomez, L. Menchi, I. Lobysheva, V. Di Mauro, H. Esfahani, G. Condorelli,
M. Leopizzi, M. Mancini, G. Zerbini, G. Condorelli, A. Stella, MiR-133a regulates collagen J.-L. Balligand, D. Catalucci, C. Dessy, MicroRNA-199a-3p and MicroRNA-199a-5p take part
1A1: Potential role of miR-133a in myocardial fibrosis in angiotensin II-dependent to a redundant network of regulation of the NOS (NO Synthase)/NO pathway
hypertension. J. Cell. Physiol. 227, 850–856 (2012). in the endothelium. Arterioscler. Thromb. Vasc. Biol. 38, 2345–2357 (2018).
123. Y. Sassi, P. Avramopoulos, D. Ramanujam, L. Grüter, S. Werfel, S. Giosele, A.-D. Brunner, 144. X. Zhao, X. Wei, X. Wang, G. Qi, Long non-coding RNA NORAD regulates angiogenesis
D. Esfandyari, A. S. Papadopoulou, B. De Strooper, N. Hübner, R. Kumarswamy, T. Thum, of human umbilical vein endothelial cells via miR-590-3p under hypoxic conditions.
X. Yin, M. Mayr, B. Laggerbauer, S. Engelhardt, Cardiac myocyte miR-29 promotes Mol. Med. Rep. 21, 2560–2570 (2020).
pathological remodeling of the heart by activating Wnt signaling. Nat. Commun. 8, 1614 145. Z.-G. Fan, X.-L. Qu, P. Chu, Y.-L. Gao, X.-F. Gao, S.-L. Chen, N.-L. Tian, MicroRNA-210
(2017). promotes angiogenesis in acute myocardial infarction. Mol. Med. Rep. 17, 5658–5665 (2018).

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


124. F. L. Xiang, M. Fang, K. E. Yutzey, Loss of -catenin in resident cardiac fibroblasts 146. N. Wang, C. Chen, D. Yang, Q. Liao, H. Luo, X. Wang, F. Zhou, X. Yang, J. Yang, C. Zeng,
attenuates fibrosis induced by pressure overload in mice. Nat. Commun. 8, 712 (2017). W. E. Wang, Mesenchymal stem cells-derived extracellular vesicles, via miR-210, improve
125. M.-H. Chen, J.-C. Liu, Y. Liu, Y.-C. Hu, X.-F. Cai, D.-C. Yin, MicroRNA-199a regulates infarcted cardiac function by promotion of angiogenesis. Biochim. Biophys. Acta - Mol.
myocardial fibrosis in rats by targeting SFRP5. Eur. Rev. Med. Pharmacol. Sci. 23, Basis Dis. 1863, 2085–2092 (2017).
3976–3983 (2019). 147. C. L. Gallant-Behm, J. Piper, B. A. Dickinson, C. M. Dalby, L. A. Pestano, A. L. Jackson,
126. J. Cui, S. Qi, R. Liao, D. Su, Y. Wang, S. Xue, MiR-574–5p promotes the differentiation A synthetic microRNA-92a inhibitor (MRG-110) accelerates angiogenesis and wound
of human cardiac fibroblasts via regulating ARID3A. Biochem. Biophys. Res. Commun. 521, healing in diabetic and nondiabetic wounds. Wound Repair Regen. 26, 311–323 (2018).
427–433 (2020). 148. A. Bonauer, G. Carmona, M. Iwasaki, M. Mione, M. Koyanagi, A. Fischer, J. Burchfield,
127. X. Yuan, J. Pan, L. Wen, B. Gong, J. Li, H. Gao, W. Tan, S. Liang, H. Zhang, X. Wang, H. Fox, C. Doebele, K. Ohtani, E. Chavakis, M. Potente, M. Tjwa, C. Urbich, A. M. Zeiher,
MiR-590-3p regulates proliferation, migration and collagen synthesis of cardiac fibroblast S. Dimmeler, MicroRNA-92a controls angiogenesis and functional recovery of ischemic
by targeting ZEB1. J. Cell. Mol. Med. 24, 227–237 (2020). tissues in Mice. Science 324, 1710–1713 (2009).
128. D. L. Brutsaert, Cardiac endothelial-myocardial signaling: Its role in cardiac growth, 149. D. Veliceasa, D. Biyashev, G. Qin, S. Misener, A. R. Mackie, R. Kishore, O. V. Volpert,
contractile performance, and rhythmicity. Physiol. Rev. 83, 59–115 (2003). Therapeutic manipulation of angiogenesis with miR-27b. Vasc. Cell 7, 6 (2015).
129. H. Falque, T. Bochaton, H. Bernelin, A. Paccalet, C. C. Da Silva, D. Baetz, E. Bonnefoy-Cudraz, 150. C.-C. Li, X.-T. Qiu, Q. Sun, J.-P. Zhou, H.-J. Yang, W.-Z. Wu, L.-F. He, C.-E. Tang, G.-G. Zhang,
N. Mewton, M. Ovize, Endothelial activation and infarct size at the acute phase Y.-P. Bai, Endogenous reduction of miR-185 accelerates cardiac function recovery in mice
of myocardial infarction. Arch. Cardiovasc. Dis. Suppl. 10, 178 (2018). following myocardial infarction via targeting of cathepsin K. J. Cell. Mol. Med. 23,
130. S. Hernández-Reséndiz, M. Muñoz-Vega, W. E. Contreras, G. E. Crespo-Avilan, 1164–1173 (2019).
J. Rodriguez-Montesinos, O. Arias-Carrión, O. Pérez-Méndez, W. A. Boisvert, K. T. Preissner, 151. A. S. Bayoumi, J.-P. Teoh, T. Aonuma, Z. Yuan, X. Ruan, Y. Tang, H. Su, N. L. Weintraub,
H. A. Cabrera-Fuentes, Responses of endothelial cells towards ischemic conditioning I.-M. Kim, MicroRNA-532 protects the heart in acute myocardial infarction, and represses
following acute myocardial infarction. Cond. Med. 1, 247–258 (2018). prss23, a positive regulator of endothelial-to-mesenchymal transition. Cardiovasc. Res.
131. M. L. Lindsey, G. P. Escobar, L. W. Dobrucki, D. K. Goshorn, S. Bouges, J. T. Mingoia, 113, 1603–1614 (2017).
D. M. McClister, H. Su, J. Gannon, C. MacGillivray, R. T. Lee, A. J. Sinusas, F. G. Spinale, 152. C. M. Bartman, Y. Oyama, K. Brodsky, L. Khailova, L. Walker, M. Koeppen, T. Eckle,
Matrix metalloproteinase-9 gene deletion facilitates angiogenesis after myocardial M. Wang, Intense light-elicited upregulation of miR-21 facilitates glycolysis
infarction. Am. J. Physiol. Heart Circ. Physiol. 290, H232–H239 (2006). and cardioprotection through Per2-dependent mechanisms. PLOS ONE 12, e0176243
132. A. Fallah, A. Sadeghinia, H. Kahroba, A. Samadi, H. R. Heidari, B. Bradaran, S. Zeinali, (2017).
O. Molavi, Therapeutic targeting of angiogenesis molecular pathways in angiogenesis- 153. R. R. Liu, J. Li, J. Y. Gong, F. Kuang, J. Y. Liu, Y. S. Zhang, Q. L. Ma, C. J. Song, A. D. Truax,
dependent diseases. Biomed. Pharmacother. 110, 775–785 (2019). F. Gao, K. Yang, B. Q. Jin, L. H. Chen, MicroRNA-141 regulates the expression level
133. Y. Zhang, W. Qin, L. Zhang, X. Wu, N. Du, Y. Hu, X. Li, N. Shen, D. Xiao, H. Zhang, Z. Li, of ICAM-1 on endothelium to decrease myocardial ischemia-reperfusion injury. Am.
Y. Zhang, H. Yang, F. Gao, Z. Du, C. Xu, B. Yang, MicroRNA-26a prevents endothelial cell J. Physiol. Circ. Physiol. 309, H1303–H1313 (2015).
apoptosis by directly targeting TRPC6 in the setting of atherosclerosis. Sci. Rep. 5, 9401 154. L. Yang, B. Wang, Q. Zhou, Y. Wang, X. Liu, Z. Liu, Z. Zhan, MicroRNA-21 prevents
(2015). excessive inflammation and cardiac dysfunction after myocardial infarction through
134. B. Qin, Y. Shu, L. Xiao, T. Lu, Y. Lin, H. Yang, Z. Lu, MicroRNA-150 targets ELK1 and modulates targeting KBTBD7. Cell Death Dis. 9, 769 (2018).
the apoptosis induced by ox-LDL in endothelial cells. Mol. Cell. Biochem. 429, 45–58 (2017). 155. S. K. Gupta, R. Itagaki, X. Zheng, S. Batkai, S. Thum, F. Ahmad, L. N. Van Aelst, A. Sharma,
135. P. Chen, J. Zhong, J. Ye, Y. He, Z. Liang, Y. Cheng, J. Zheng, H. Chen, C. Chen, miR-324-5p M.-T. Piccoli, F. Weinberger, J. Fiedler, M. Heuser, S. Heymans, C. S. Falk, R. Förster,
protects against oxidative stress-induced endothelial progenitor cell injury by targeting S. Schrepfer, T. Thum, miR-21 promotes fibrosis in an acute cardiac allograft
Mtfr1. J. Cell. Physiol. 234, 22082–22092 (2019). transplantation model. Cardiovasc. Res. 110, 215–226 (2016).
136. G. Ghosh, I. V. Subramanian, N. Adhikari, X. Zhang, H. P. Joshi, D. Basi, 156. J. Li, S. X. X. Cai, Q. He, H. Zhang, D. Friedberg, F. Wang, A. N. Redington, Intravenous
Y. S. Chandrashekhar, J. L. Hall, S. Roy, Y. Zeng, S. Ramakrishnan, Hypoxia-induced miR-144 reduces left ventricular remodeling after myocardial infarction. Basic Res.
microRNA-424 expression in human endothelial cells regulates HIF- isoforms Cardiol. 113, 36 (2018).
and promotes angiogenesis. J. Clin. Invest. 120, 4141–4154 (2010). 157. H.-M. Xu, F.-H. Sui, M.-H. Sun, G.-L. Guo, Downregulated microRNA-224 aggravates
137. G.-H. Dai, P.-Z. Ma, X.-B. Song, N. Liu, T. Zhang, B. Wu, MicroRNA-223-3p inhibits vulnerable atherosclerotic plaques and vascular remodeling in acute coronary syndrome
the angiogenesis of ischemic cardiac microvascular endothelial cells via affecting through activation of the TGF-/Smad pathway. J. Cell. Physiol. 234, 2537–2551 (2019).
RPS6KB1/hif-1a signal pathway. PLOS ONE 9, e108468 (2014). 158. J. Yang, M. E. Brown, H. Zhang, M. Martinez, Z. Zhao, S. Bhutani, S. Yin, D. Trac, J. J. Xi,
138. J. Fiedler, V. Jazbutyte, B. C. Kirchmaier, S. K. Gupta, J. Lorenzen, D. Hartmann, M. E. Davis, High-throughput screening identifies microRNAs that target Nox2
P. Galuppo, S. Kneitz, J. T. G. Pena, C. Sohn-Lee, X. Loyer, J. Soutschek, T. Brand, T. Tuschl, and improve function after acute myocardial infarction. Am. J. Physiol. Hear. Circ. Physiol.
J. Heineke, U. Martin, S. Schulte-Merker, G. Ertl, S. Engelhardt, J. Bauersachs, T. Thum, 312, H1002–H1012 (2017).
MicroRNA-24 regulates vascularity after myocardial infarction. Circulation 124, 720–730 159. G. de Couto, R. Gallet, L. Cambier, E. Jaghatspanyan, N. Makkar, J. F. Dawkins,
(2011). B. P. Berman, E. Marbán, Exosomal microRNA transfer into macrophages mediates
139. S. Wang, A. B. Aurora, B. A. Johnson, X. Qi, J. M. Anally, J. A. Hill, J. A. Richardson, cellular postconditioning. Circulation 136, 200–214 (2017).
R. Bassel-Duby, E. N. Olson, The endothelial-specific microRNA miR-126 governs vascular 160. V. N. S. Garikipati, S. K. Verma, D. Jolardarashi, Z. Cheng, J. Ibetti, M. Cimini, Y. Tang,
integrity and angiogenesis. Dev. Cell 15, 261–271 (2008). M. Khan, Y. Yue, C. Benedict, E. Nickoloff, M. M. Truongcao, E. Gao, P. Krishnamurthy,
140. T. Ma, Y. Chen, Y. Chen, Q. Meng, J. Sun, L. Shao, Y. Yu, H. Huang, Y. Hu, Z. Yang, J. Yang, D. A. Goukassian, W. J. Koch, R. Kishore, Therapeutic inhibition of miR-375 attenuates
Z. Shen, MicroRNA-132, delivered by mesenchymal stem cell-derived exosomes, post-myocardial infarction inflammatory response and left ventricular dysfunction via
promote angiogenesis in myocardial infarction. Stem Cells Int. 2018, 3290372 (2018). PDK-1-AKT signalling axis. Cardiovasc. Res. 113, 938–949 (2017).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 17 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

161. L. Shao, Y. Zhang, X. Pan, B. Liu, C. Liang, Y. Zhang, Y. Wang, B. Yan, W. Xie, Y. Sun, Z. Shen, 183. K. A. Lennox, M. A. Behlke, Chemical modification and design of anti-miRNA
X.-Y. Yu, Y. Li, Knockout of beta-2 microglobulin enhances cardiac repair by modulating oligonucleotides. Gene Ther. 18, 1111–1120 (2011).
exosome imprinting and inhibiting stem cell-induced immune rejection. Cell. Mol. Life Sci. 184. G. Meister, M. Landthaler, Y. Dorsett, T. Tuschl, Sequence-specific inhibition of
77, 937–952 (2020). microRNA-and siRNA-induced RNA silencing. RNA 10, 544–550 (2004).
162. J. Zhao, X. Li, J. Hu, F. Chen, S. Qiao, X. Sun, L. Gao, J. Xie, B. Xu, Mesenchymal stromal 185. J. Krützfeldt, N. Rajewsky, R. Braich, K. G. Rajeev, T. Tuschl, M. Manoharan, M. Stoffel,
cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through Silencing of microRNAs in vivo with “antagomirs”. Nature 438, 685–689 (2005).
miR-182-regulated macrophage polarization. Cardiovasc. Res. 115, 1205–1216 (2019). 186. T. A. Vickers, J. R. Wyatt, T. Burckin, C. F. Bennett, S. M. Freier, Fully modified 2′ MOE
163. T. Bejerano, S. Etzion, S. Elyagon, Y. Etzion, S. Cohen, Nanoparticle delivery of miRNA-21 oligonucleotides redirect polyadenylation. Nucleic Acids Res. 29, 1293–1299 (2001).
mimic to cardiac macrophages improves myocardial remodeling after myocardial 187. J. Elmén, M. Lindow, A. Silahtaroglu, M. Bak, M. Christensen, A. Lind-Thomsen,
infarction. Nano Lett. 18, 5885–5891 (2018). M. Hedtjärn, J. B. Hansen, H. F. Hansen, E. M. Straarup, K. M. Cullagh, P. Kearney,
164. Z. M. Ye, S. Yang, Y.-p. Xia, R.-t. Hu, S. Chen, B.-w. Li, S.-l. Chen, X.-y. Luo, L. Mao, Y. Li, S. Kauppinen, Antagonism of microRNA-122 in mice by systemically administered
H. Jin, C. Qin, B. Hu, LncRNA MIAT sponges miR-149-5p to inhibit efferocytosis LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver.
in advanced atherosclerosis through CD47 upregulation. Cell Death Dis. 10, 138 (2019). Nucleic Acids Res. 36, 1153–1162 (2008).
165. M. Arif, R. Pandey, P. Alam, S. Jiang, S. Sadayappan, A. Paul, R. P. H. Ahmed, MicroRNA- 188. S. Obad, C. O. dos Santos, A. Petri, M. Heidenblad, O. Broom, C. Ruse, C. Fu, M. Lindow,
210-mediated proliferation, survival, and angiogenesis promote cardiac repair post J. Stenvang, E. M. Straarup, H. F. Hansen, T. Koch, D. Pappin, G. J. Hannon, S. Kauppinen,
myocardial infarction in rodents. J. Mol. Med. 95, 1369–1385 (2017). Silencing of microRNA families by seed-targeting tiny LNAs. Nat. Genet. 43, 371–380 (2011).
166. S. Hu, M. Huang, Z. Li, F. Jia, Z. Ghosh, M. A. Lijkwan, P. Fasanaro, N. Sun, X. Wang, 189. M. S. Ebert, J. R. Neilson, P. A. Sharp, MicroRNA sponges: Competitive inhibitors of small

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


F. Martelli, R. C. Robbins, J. C. Wu, MicroRNA-210 as a novel therapy for treatment RNAs in mammalian cells. Nat. Methods 4, 721–726 (2007).
of ischemic heart disease. Circulation 122, S124–S131 (2010). 190. M. S. Ebert, P. A. Sharp, MicroRNA sponges: Progress and possibilities. RNA 16, 2043–2050
167. L. Huang, B. Guo, S. Liu, C. Miao, Y. Li, Inhibition of the LncRNA Gpr19 attenuates (2010).
ischemia-reperfusion injury after acute myocardial infarction by inhibiting apoptosis 191. E. C. Keeley, J. A. Boura, C. L. Grines, Primary angioplasty versus intravenous thrombolytic
and oxidative stress via the miR-324-5p/Mtfr1 axis. IUBMB Life 72, 373–383 (2020). therapy for acute myocardial infarction: A quantitative review of 23 randomised trials.
168. X. Wang, X. Zhang, X.-P. Ren, J. Chen, H. Liu, J. Yang, M. Medvedovic, Z. Hu, G.-C. Fan, Lancet 361, 13–20 (2003).
MicroRNA-494 targeting both proapoptotic and antiapoptotic proteins protects against 192. A. Foinquinos, S. Batkai, C. Genschel, J. Viereck, S. Rump, M. Gyöngyösi, D. Traxler,
ischemia/reperfusion-induced cardiac injury. Circulation 122, 1308–1318 (2010). M. Riesenhuber, A. Spannbauer, D. Lukovic, N. Weber, K. Zlabinger, E. Hašimbegović,
169. Q. Su, X.-W. Lv, Y.-H. Sun, Z.-L. Ye, B.-H. Kong, Z.-B. Qin, MicroRNA-494 inhibits the LRG1 J. Winkler, J. Fiedler, S. Dangwal, M. Fischer, J. de la Roche, D. Wojciechowski, T. Kraft,
expression to induce proliferation and migration of VECs in rats following myocardial R. Garamvölgyi, S. Neitzel, S. Chatterjee, X. Yin, C. Bär, M. Mayr, K. Xiao, T. Thum,
infarction. Mol. Ther. Nucleic Acids 18, 110–122 (2019). Preclinical development of a miR-132 inhibitor for heart failure treatment. Nat. Commun.
170. H. el Azzouzi, S. Leptidis, E. Dirkx, J. Hoeks, B. van Bree, K. Brand, E. A. McClellan, E. Poels, 11, 633 (2020).
J. C. Sluimer, M. M. G. van den Hoogenhof, A. S. Armand, X. Yin, S. Langley, M. Bourajjaj, 193. S. F. Dowdy, Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 35,
S. Olieslagers, J. Krishnan, M. Vooijs, H. Kurihara, A. Stubbs, Y. M. Pinto, W. Krek, M. Mayr, 222–229 (2017).
P. A. D. C. Martins, P. Schrauwen, L. J. De Windt, The hypoxia-inducible microRNA cluster 194. J. A. Kulkarni, M. M. Darjuan, J. E. Mercer, S. Chen, R. van der Meel, J. L. Thewalt,
miR-199a∼214 targets myocardial PPAR and impairs mitochondrial fatty acid oxidation. Y. Y. C. Tam, P. R. Cullis, On the formation and morphology of lipid nanoparticles
Cell Metab. 18, 341–354 (2013). containing ionizable cationic lipids and sirna. ACS Nano 12, 4787–4795 (2018).
171. Z. Li, L. Liu, N. Hou, Y. Song, X. An, Y. Zhang, X. Yang, J. Wang, miR-199-sponge transgenic 195. H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J. J. Lee, J. O. Lötvall, Exosome-mediated
mice develop physiological cardiac hypertrophy. Cardiovasc. Res. 110, 258–267 (2016). transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between
172. K.-m. Park, J.-p. Teoh, Y. Wang, Z. Broskova, A. S. Bayoumi, Y. Tang, H. Su, N. L. Weintraub, cells. Nat. Cell Biol. 9, 654–659 (2007).
I.-m. Kim, Carvedilol-responsive microRNAs, miR-199a-3p and -214 protect 196. S. Bian, L. Zhang, L. Duan, X. Wang, Y. Min, H. Yu, Extracellular vesicles derived
cardiomyocytes from simulated ischemia-reperfusion injury. Am. J. Physiol. Circ. Physiol. from human bone marrow mesenchymal stem cells promote angiogenesis in a rat
311, H371–H383 (2016). myocardial infarction model. J. Mol. Med. (Berl) 92, 387–397 (2014).
173. L. Chen, F.-Y. Wang, Z.-Y. Zeng, L. Cui, J. Shen, X.-W. Song, P. Li, X.-X. Zhao, Y.-W. Qin, 197. B. Liu, B. W. Lee, K. Nakanishi, A. Villasante, R. Williamson, J. Metz, J. Kim, M. Kanai, L. Bi,
MicroRNA-199a acts as a potential suppressor of cardiomyocyte autophagy through K. Brown, G. Di Paolo, S. Homma, P. A. Sims, V. K. Topkara, G. Vunjak-Novakovic, Cardiac
targeting Hspa5. Oncotarget 8, 63825–63834 (2017). recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes
174. R. Hinkel, D. Ramanujam, V. Kaczmarek, A. Howe, K. Klett, C. Beck, A. Dueck, T. Thum, derived from induced pluripotent stem cells. Nat. Biomed. Eng. 2, 293–303 (2018).
K. L. Laugwitz, L. Maegdefessel, C. Weber, C. Kupatt, S. Engelhardt, AntimiR-21 prevents 198. R. Gallet, J. Dawkins, J. Valle, E. Simsolo, G. de Couto, R. Middleton, E. Tseliou,
myocardial dysfunction in a pig model of ischemia/reperfusion injury. J. Am. Coll. Cardiol. D. Luthringer, M. Kreke, R. R. Smith, L. Marbán, B. Ghaleh, E. Marbán, Exosomes secreted
75, 1788–1800 (2020). by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling,
175. J. Hu, C.-X. Huang, P.-P. Rao, J.-P. Zhou, X. Wang, L. Tang, M.-X. Liu, G.-G. Zhang, Inhibition and improve function in acute and chronic porcine myocardial infarction. Eur. Heart J. 38,
of microRNA-155 attenuates sympathetic neural remodeling following myocardial 201–211 (2017).
infarction via reducing M1 macrophage polarization and inflammatory responses 199. K. Pachler, T. Lener, D. Streif, Z. A. Dunai, A. Desgeorges, M. Feichtner, M. Öller,
in mice. Eur. J. Pharmacol. 851, 122–132 (2019). K. Schallmoser, E. Rohde, M. Gimona, A good manufacturing practice–grade standard
176. Y. Ye, Z. Hu, Y. Lin, C. Zhang, J. R. Perez-Polo, Downregulation of microRNA-29 by protocol for exclusively human mesenchymal stromal cell–derived extracellular vesicles.
antisense inhibitors and a PPAR- agonist protects against myocardial ischaemia– Cytotherapy 19, 458–472 (2017).
reperfusion injury. Cardiovasc. Res. 87, 535–544 (2010). 200. Y. Chen, Y. Zhao, W. Chen, L. Xie, Z.-A. Zhao, J. Yang, Y. Chen, W. Lei, Z. Shen,
177. Z.-X. Guo, F.-Z. Zhou, W. Song, L.-L. Yu, W.-J. Yan, L.-H. Yin, H. Sang, H.-Y. Zhang, MicroRNA-133 overexpression promotes the therapeutic efficacy of mesenchymal stem
Suppression of microRNA-101 attenuates hypoxia-induced myocardial H9c2 cell injury cells on acute myocardial infarction. Stem Cell Res. Ther. 8, 268 (2017).
by targeting DIMT1-Sp1/survivin pathway. Eur. Rev. Med. Pharmacol. Sci. 22, 6965–6976 201. M. Morishita, Y. Takahashi, M. Nishikawa, Y. Takakura, Pharmacokinetics of exosomes—an
(2018). important factor for elucidating the biological roles of exosomes
178. C. Wang, C. Zhang, L. Liu, A. Xi, B. Chen, Y. Li, J. Du, Macrophage-derived mir-155- and for the development of exosome-based therapeutics. J. Pharm. Sci. 106, 2265–2269
containing exosomes suppress fibroblast proliferation and promote fibroblast (2017).
inflammation during cardiac injury. Mol. Ther. 25, 192–204 (2017). 202. A. Wittrup, A. Ai, X. Liu, P. Hamar, R. Trifonova, K. Charisse, M. Manoharan, T. Kirchhausen,
179. J. Guo, H.-B. Liu, C. Sun, X.-Q. Yan, J. Hu, J. Yu, Y. Yuan, Z.-M. Du, MicroRNA-155 promotes J. Lieberman, Visualizing lipid-formulated siRNA release from endosomes and target
myocardial infarction-induced apoptosis by targeting RNA-binding protein QKI. gene knockdown. Nat. Biotechnol. 33, 870–876 (2015).
Oxid. Med. Cell. Longev. 2019, 4579806 (2019). 203. A. D. Springer, S. F. Dowdy, GalNAc-siRNA conjugates: Leading the way for delivery
180. J. Hanna, G. S. Hossain, J. Kocerha, The potential for microRNA therapeutics and clinical of RNAi therapeutics. Nucleic Acid Ther. 28, 109–118 (2018).
research. Front. Genet. 10, 478 (2019). 204. K. I. Mentkowski, J. K. Lang, Exosomes engineered to express a cardiomyocyte binding
181. R. L. Juliano, The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 44, peptide demonstrate improved cardiac retention in vivo. Sci. Rep. 9, 10041 (2019).
6518–6548 (2016). 205. X. Xue, X. Shi, H. Dong, S. You, H. Cao, K. Wang, Y. Wen, D. Shi, B. He, Y. Li, Delivery
182. F. Eckstein, Phosphorothioates, essential components of therapeutic oligonucleotides. of microRNA-1 inhibitor by dendrimer-based nanovector: An early targeting therapy
Nucleic Acid Ther. 24, 374–387 (2014). for myocardial infarction in mice. Nanomedicine 14, 619–631 (2018).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 18 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

206. T. Sugo, M. Terada, T. Oikawa, K. Miyata, S. Nishimura, E. Kenjo, M. Ogasawara-Shimizu, 229. L. Roca-Alonso, L. Castellano, A. Mills, A. F. Dabrowska, M. B. Sikkel, L. Pellegrino, J. Jacob,
Y. Makita, S. Imaichi, S. Murata, K. Otake, K. Kikuchi, M. Teratani, Y. Masuda, T. Kamei, A. E. Frampton, J. Krell, R. C. Coombes, S. E. Harding, A. R. Lyon, J. Stebbing, Myocardial
S. Takagahara, S. Ikeda, T. Ohtaki, H. Matsumoto, Development of antibody-siRNA MiR-30 downregulation triggered by doxorubicin drives alterations in -adrenergic
conjugate targeted to cardiac and skeletal muscles. J. Control. Release 237, 1–13 (2016). signaling and enhances apoptosis. Cell Death Dis. 6, e1754 (2015).
207. M. Liu, M. Li, S. Sun, B. Li, D. Du, J. Sun, F. Cao, H. Li, F. Jia, T. Wang, N. Chang, H. Yu, 230. A. B. Aurora, A. I. Mahmoud, X. Luo, B. A. Johnson, E. van Rooij, S. Matsuzaki,
Q. Wang, H. Peng, The use of antibody modified liposomes loaded with AMO-1 to deliver K. M. Humphries, J. A. Hill, R. Bassel-Duby, H. A. Sadek, E. N. Olson, MicroRNA-214 protects
oligonucleotides to ischemic myocardium for arrhythmia therapy. Biomaterials 35, the mouse heart from ischemic injury by controlling Ca2+ overload and cell death. J. Clin.
3697–3707 (2014). Invest. 122, 1222–1232 (2012).
208. T. L. Cuellar, D. Barnes, C. Nelson, J. Tanguay, S.-F. Yu, X. Wen, S. J. Scales, J. Gesch, 231. Y. Hao, H. Yuan, H. Yu, Downregulation of miR-483-5p decreases hypoxia-induced injury
D. Davis, A. van Brabant Smith, D. Leake, R. Vandlen, C. W. Siebel, Systematic evaluation in human cardiomyocytes by targeting MAPK3. Cell. Mol. Biol. Lett. 25, 20 (2020).
of antibody-mediated siRNA delivery using an industrial platform of THIOMAB-siRNA 232. X.-P. Ren, J. Wu, X. Wang, M. A. Sartor, J. Qian, K. Jones, P. Nicolaou, T. J. Pritchard,
conjugates. Nucleic Acids Res. 43, 1189–1203 (2015). G.-C. Fan, MicroRNA-320 is involved in the regulation of cardiac ischemia/reperfusion
209. R. L. Juliano, X. Ming, K. Carver, B. Laing, Cellular uptake and intracellular trafficking injury by targeting heat-shock protein 20. Circulation 119, 2357–2366 (2009).
of oligonucleotides: Implications for oligonucleotide pharmacology. Nucleic Acid Ther. 24, 233. Y. Shen, Z. Shen, L. Miao, X. Xin, S. Lin, Y. Zhu, W. Guo, Y. Z. Zhu, miRNA-30 family
101–113 (2014). inhibition protects against cardiac ischemic injury by regulating cystathionine--lyase
210. P. Lönn, A. D. Kacsinta, X. S. Cui, A. S. Hamil, M. Kaulich, K. Gogoi, S. F. Dowdy, Enhancing expression. Antioxid. Redox Signal. 22, 224–240 (2015).
endosomal escape for intracellular delivery of macromolecular biologic therapeutics. 234. X. Li, Z. Zeng, Q. Li, Q. Xu, J. Xie, H. Hao, G. Luo, W. Liao, J. Bin, X. Huang, Y. Liao, Inhibition

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


Sci. Rep. 6, 32301 (2016). of microRNA-497 ameliorates anoxia/reoxygenation injury in cardiomyocytes by
211. J. Gilleron, W. Querbes, A. Zeigerer, A. Borodovsky, G. Marsico, U. Schubert, K. Manygoats, suppressing cell apoptosis and enhancing autophagy. Oncotarget 6, 18829–18844 (2015).
S. Seifert, C. Andree, M. Stöter, H. Epstein-Barash, L. Zhang, V. Koteliansky, K. Fitzgerald, 235. G. Wu, J. Tan, J. Li, X. Sun, L. Du, S. Tao, miRNA-145-5p induces apoptosis after
E. Fava, M. Bickle, Y. Kalaidzidis, A. Akinc, M. Maier, M. Zerial, Image-based analysis of lipid ischemia-reperfusion by targeting dual specificity phosphatase 6. J. Cell. Physiol. 234,
nanoparticle-mediated siRNA delivery, intracellular trafficking and endosomal escape. 16281–16289 (2019).
Nat. Biotechnol. 31, 638–646 (2013). 236. Y. Liu, X.-M. Qian, Q.-C. He, J.-K. Weng, MiR-421 inhibition protects H9c2 cells against
212. M. Dominska, D. M. Dykxhoorn, Breaking down the barriers: siRNA delivery hypoxia/reoxygenation-induced oxidative stress and apoptosis by targeting Sirt3.
and endosome escape. J. Cell Sci. 123, 1183–1189 (2010). Perfusion 35, 255–262 (2019).
213. M. Stessl, C. R. Noe, J. Winkler, in From Nucleic Acids Sequences to Molecular Medicine, 237. B. Long, K. Wang, N. Li, I. Murtaza, J.-Y. Xiao, Y.-Y. Fan, C.-Y. Liu, W.-H. Li, Z. Cheng, P. F. Li,
V. A. Erdmann, J. Barciszewski, Eds. (Springer Berlin Heidelberg, 2012), pp. 67–83. MiR-761 regulates the mitochondrial network by targeting mitochondrial fission factor.
214. J. Winkler, M. Stessl, J. Amartey, C. R. Noe, Off-target effects related Free Radic. Biol. Med. 65, 371–379 (2013).
to the phosphorothioate modification of nucleic acids. ChemMedChem 5, 1344–1352 238. X. Wang, T. Ha, J. Zou, D. Ren, L. Liu, X. Zhang, J. Kalbfleisch, X. Gao, D. Williams, C. Li,
(2010). MicroRNA-125b protects against myocardial ischaemia/reperfusion injury via
215. M. Sioud, Recent advances in small interfering RNA sensing by the immune system. targeting p53-mediated apoptotic signalling and TRAF6. Cardiovasc. Res. 102,
N. Biotechnol. 27, 236–242 (2010). 385–395 (2014).
216. N. Pencheva, H. Tran, C. Buss, D. Huh, M. Drobnjak, K. Busam, S. F. Tavazoie, Convergent 239. J. Pan, M. Alimujiang, Q. Chen, H. Shi, X. Luo, Exosomes derived from miR-146a-modified
multi-miRNA targeting of ApoE grives LRP1/LRP8-dependent melanoma metastasis adipose-derived stem cells attenuate acute myocardial infarction−induced myocardial
and angiogenesis. Cell 151, 1068–1082 (2012). damage via downregulation of early growth response factor 1. J. Cell. Biochem. 120,
217. Z. Wang, X. Ma, Q. Cai, X. Wang, B. Yu, Q. Cai, B. Liu, Z. Zhu, C. Li, MiR-199a-3p promotes 4433–4443 (2019).
gastric cancer progression by targeting ZHX1. FEBS Lett. 588, 4504–4512 (2014). 240. X. Wang, T. Ha, L. Liu, J. Zou, X. Zhang, J. Kalbfleisch, X. Gao, D. Williams, C. Li, Increased
218. B. C. Bernardo, X.-M. Gao, C. E. Winbanks, E. J. H. Boey, Y. K. Tham, H. Kiriazis, expression of microRNA-146a decreases myocardial ischaemia/reperfusion injury.
P. Gregorevic, S. Obad, S. Kauppinen, X.-J. Du, R. C. Y. Lin, J. R. McMullen, Therapeutic Cardiovasc. Res. 97, 432–442 (2013).
inhibition of the miR-34 family attenuates pathological cardiac remodeling and improves 241. S. Zhang, R. Zhang, F. Wu, X. Li, MicroRNA-208a regulates H9c2 cells simulated
heart function. Proc. Natl. Acad. Sci. U.S.A. 109, 17615–17620 (2012). ischemia-reperfusion myocardial injury via targeting CHD9 through Notch/NF-kappa B
219. N. H. Kim, H. S. Kim, N.-G. Kim, I. Lee, H.-S. Choi, X.-Y. Li, S. E. Kang, S. Y. Cha, J. K. Ryu, signal pathways. Int. Heart. J. 59, 580–588 (2018).
J. M. Na, C. Park, K. Kim, S. Lee, B. M. Gumbiner, J. I. Yook, S. J. Weiss, p53 and microRNA-34 242. W. Huang, S.-S. Tian, P.-Z. Hang, C. Sun, J. Guo, Z.-M. Du, Combination of microRNA-21
are suppressors of canonical Wnt signaling. Sci. Signal. 4, ra71 (2011). and microRNA-146a attenuates cardiac dysfunction and apoptosis during acute
220. L. Zhang, Y. Liao, L. Tang, MicroRNA-34 family: A potential tumor suppressor myocardial infarction in mice. Mol. Ther. Nucleic Acids 5, e296 (2016).
and therapeutic candidate in cancer. J. Exp. Clin. Cancer Res. 38, 53 (2019). 243. L. Huang, L. Yang, Y. Ding, X. Jiang, Z. Xia, Z. You, Human umbilical cord mesenchymal
221. A. Mathur, P. Loskill, K. Shao, N. Huebsch, S. G. Hong, S. G. Marcus, N. Marks, M. Mandegar, stem cells-derived exosomes transfers microRNA-19a to protect cardiomyocytes
B. R. Conklin, L. P. Lee, K. E. Healy, Human iPSC-based cardiac microphysiological system from acute myocardial infarction by targeting SOX6. Cell Cycle 19, 339–353 (2020).
for drug screening applications. Sci. Rep. 5, 8883 (2015). 244. J. Ma, Z. Chen, Y. Ma, Y. Xia, K. Hu, Y. Zhou, A. Chen, J. Qian, J. Ge, MicroRNA-19a
222. K. R. Chi, Revolution dawning in cardiotoxicity testing. Nat. Rev. Drug Discov. 12, 565–567 attenuates hypoxia-induced cardiomyocyte apoptosis by downregulating NHE-1
(2013). expression and decreasing calcium overload. J. Cell. Biochem. 121, 1747–1758 (2020).
223. P. W. Burridge, E. Matsa, P. Shukla, Z. C. Lin, J. M. Churko, A. D. Ebert, F. Lan, S. Diecke, 245. Q. Yang, K. Yang, A. Li, microRNA-21 protects against ischemia-reperfusion and hypoxia-
B. Huber, N. M. Mordwinkin, J. R. Plews, O. J. Abilez, B. Cui, J. D. Gold, J. C. Wu, Chemically reperfusion- induced cardiocyte apoptosis via the phosphatase and tensin homolog/
defined generation of human cardiomyocytes. Nat. Methods 11, 855–860 (2014). Akt- dependent mechanism. Mol. Med. Rep. 9, 2213–2220 (2014).
224. K. Ronaldson-Bouchard, S. P. Ma, K. Yeager, T. Chen, L. J. Song, D. Sirabella, K. Morikawa, 246. Y. Jin, S. Ni, miR-496 remedies hypoxia reoxygenation–induced H9c2 cardiomyocyte
D. Teles, M. Yazawa, G. Vunjak-Novakovic, Advanced maturation of human cardiac tissue apoptosis via Hook3-targeted PI3k/Akt/mTOR signaling pathway activation. J. Cell.
grown from pluripotent stem cells. Nature 556, 239–243 (2018). Biochem. 121, 698–712 (2020).
225. N. Huebsch, P. Loskill, N. Deveshwar, C. I. Spencer, L. M. Judge, M. A. Mandegar, C. B. Fox, 247. Y. Shi, Y. Han, L. Niu, J. Li, Y. Chen, MiR-499 inhibited hypoxia/reoxygenation induced
T. M. A. Mohamed, Z. Ma, A. Mathur, A. M. Sheehan, A. Truong, M. Saxton, J. Yoo, cardiomyocytes injury by targeting SOX6. Biotechnol. Lett. 41, 837–847 (2019).
D. Srivastava, T. A. Desai, P.-L. So, K. E. Healy, B. R. Conklin, Miniaturized iPS-cell-derived 248. Q. Li, J. Xie, R. Li, J. Shi, J. Sun, R. Gu, L. Ding, L. Wang, B. Xu, Overexpression of microRNA-
cardiac muscles for physiologically relevant drug response analyses. Sci. Rep. 6, 24726 99a attenuates heart remodelling and improves cardiac performance after myocardial
(2016). infarction. J. Cell. Mol. Med. 18, 919–928 (2014).
226. J. Pellman, J. Zhang, F. Sheikh, Myocyte-fibroblast communication in cardiac fibrosis 249. L.-C. Gong, H.-M. Xu, G.-L. Guo, T. Zhang, J.-W. Shi, C. Chang, Long non-coding RNA H19
and arrhythmias: Mechanisms and model systems. J. Mol. Cell. Cardiol. 94, 22–31 (2016). protects H9c2 cells against hypoxia-induced injury by targeting MicroRNA-139.
227. G. Gintant, P. Burridge, L. Gepstein, S. Harding, T. Herron, C. Hong, J. Jalife, J. C. Wu, Use Cell. Physiol. Biochem. 44, 857–869 (2017).
of human induced pluripotent stem cell-derived cardiomyocytes in preclinical cancer 250. K. Shi, H. Sun, H. Zhang, D. Xie, B. Yu, MiR-34a-5p aggravates hypoxia-induced apoptosis
drug cardiotoxicity testing: A scientific statement from the american heart association. by targeting ZEB1 in cardiomyocytes. Biol. Chem. 400, 227–236 (2019).
Circ. Res. 125, e75–e92 (2019). 251. C. Liu, M. Tang, X. Zhang, J. Li, G. Cao, Knockdown of miR-665 protects against
228. Y. Dai, T. Yan, Y. Gao, Silence of miR-32-5p promotes endothelial cell viability by targeting cardiomyocyte ischemia/reperfusion injury-induced ROS accumulation and apoptosis
KLF2 and serves as a diagnostic biomarker of acute myocardial infarction. Diagn. Pathol. through the activation of Pak1/Akt signaling in myocardial infarction. Int. Heart J. 61,
15, 19 (2020). 347–354 (2020).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 19 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

252. S. Rane, M. He, D. Sayed, H. Vashistha, A. Malhotra, J. Sadoshima, D. E. Vatner, S. F. Vatner, 274. A. Ruiz-Velasco, M. Zi, S. S. Hille, T. Azam, N. Kaur, J. Jiang, B. Nguyen, K. Sekeres, P. Binder,
M. Abdellatif, Downregulation of MiR-199a derepresses hypoxia-inducible factor-1 L. Collins, F. Pu, H. Xiao, K. Guan, N. Frey, E. J. Cartwright, O. J. Müller, X. Wang, W. Liu,
and sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ. Res. Targeting mir128-3p alleviates myocardial insulin resistance and prevents ischemia-
104, 879–886 (2009). induced heart failure. eLife 9, e54298 (2020).
253. J.-Y. Zhang, Z. Yang, K. Fang, Z.-L. Shi, D.-H. Ren, J. Sun, Long noncoding RNA ILF3-AS1 275. M. Khan, E. Nickoloff, T. Abramova, J. Johnson, S. K. Verma, P. Krishnamurthy, A. R. Mackie,
regulates myocardial infarction via the miR-212-3p/SIRT1 axis and PI3K/Akt signaling E. Vaughan, V. N. S. Garikipati, C. Benedict, V. Ramirez, E. Lambers, A. Ito, E. Gao,
pathway. Eur. Rev. Med. Pharmacol. Sci. 24, 2647–2658 (2020). S. Misener, T. Luongo, J. Elrod, G. Qin, S. R. Houser, W. J. Koch, R. Kishore, Embryonic stem
254. X. Wang, B. Yuan, B. Cheng, Y. Liu, B. Zhang, X. Wang, X. Lin, B. Yang, G. Gong, Crocin cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac
alleviates myocardial ischemia/reperfusion-induced endoplasmic reticulum stress via function following myocardial infarction. Circ. Res. 117, 52–64 (2015).
regulation of miR-34a/Sirt1/Nrf2 pathway. Shock 51, 123–130 (2019). 276. S. Deng, Q. Zhao, L. Zhen, C. Zhang, C. Liu, G. Wang, L. Zhang, L. Bao, Y. Lu, L. Meng, J. Lü,
255. L.-P. Zhu, T. Tian, J.-Y. Wang, J.-N. He, T. Chen, M. Pan, L. Xu, H.-X. Zhang, X.-T. Qiu, C.-C. Li, P. Yu, X. Lin, Y. Zhang, Y.-H. Chen, H. Fan, W. C. Cho, Z. Liu, Z. Yu, Neonatal heart-enriched
K.-K. Wang, H. Shen, G.-G. Zhang, Y.-P. Bai, Hypoxia-elicited mesenchymal stem miR-708 promotes proliferation and stress resistance of cardiomyocytes in rodents.
cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention Theranostics 7, 1953–1965 (2017).
of cell death in myocardial infarction. Theranostics 8, 6163–6177 (2018). 277. J. Chen, Z.-P. Huang, H. Y. Seok, J. Ding, M. Kataoka, Z. Zhang, X. Hu, G. Wang, Z. Lin,
256. T. Zhou, G. Qin, L. Yang, D. Xiang, S. Li, LncRNA XIST regulates myocardial infarction by S. Wang, W. T. Pu, R. Liao, D.-Z. Wang, Mir-17-92 induce cardiomyocyte proliferation
targeting miR-130a-3p. J. Cell. Physiol. 234, 8659–8667 (2019). in postnatal and adult hearts, 2013. Circ. Res. 112, 1557–1566 (2013).
257. Z. Zhao, S. Du, S. Shen, L. Wang, microRNA-132 inhibits cardiomyocyte apoptosis 278. M. Yang, D.-Y. Kong, J.-C. Chen, Inhibition of miR-148b ameliorates myocardial ischemia/

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


and myocardial remodeling in myocardial infarction by targeting IL-1. J. Cell. Physiol. reperfusion injury via regulation of Wnt/-catenin signaling pathway. J. Cell. Physiol. 234,
235, 2710–2721 (2020). 17757–17766 (2019).
258. Y.-L. Pan, Z.-Y. Han, S.-F. He, W. Yang, J. Cheng, Y. Zhang, Z.-W. Chen, MiR-133b-5p 279. R. Pandey, S. Velasquez, S. Durrani, M. Jiang, M. Neiman, J. S. Crocker, J. B. Benoit,
contributes to hypoxic preconditioning-mediated cardioprotection by inhibiting J. Rubinstein, A. Paul, R. P. H. Ahmed, MicroRNA-1825 induces proliferation of adult
the activation of caspase-8 and caspase-3 in cardiomyocytes. Mol. Med. Rep. 17, cardiomyocytes and promotes cardiac regeneration post ischemic injury. Am. J. Transl.
7097–7104 (2018). Res. 9, 3120–3137 (2017).
259. S. Yang, H. Li, L. Chen, MicroRNA-140 attenuates myocardial ischemia-reperfusion injury 280. F. Gao, M. Kataoka, N. Liu, T. Liang, Z.-P. Huang, F. Gu, J. Ding, J. Liu, F. Zhang, Q. Ma,
through suppressing mitochondria-mediated apoptosis by targeting YES1. J. Cell. Y. Wang, M. Zhang, X. Hu, J. Kyselovic, X. Hu, W. T. Pu, J. Wang, J. Chen, D.-Z. Wang,
Biochem. 120, 3813–3821 (2019). Therapeutic role of miR-19a/19b in cardiac regeneration and protection from myocardial
260. T. G. Hullinger, R. L. Montgomery, A. G. Seto, B. A. Dickinson, H. M. Semus, J. M. Lynch, infarction. Nat. Commun. 10, 1802 (2019).
C. M. Dalby, K. Robinson, C. Stack, P. A. Latimer, J. M. Hare, E. N. Olson, E. van Rooij, 281. A. Aguirre, N. Montserrat, S. Zachiggna, E. Nivet, T. Hishida, M. N. Krause, L. Kurian, A. Ocampo,
Inhibition of miR-15 protects against cardiac ischemic injury. Circ. Res. 110, 71–81 E. Vazquez-Ferrer, C. Rodriguez-Esteban, S. Kumar, J. J. Moresco, J. R. Yates III, J. M. Campistol,
(2012). I. Sancho-Martinez, M. Giacca, J. C. I. Belmonte, In vivo activation of a conserved
261. Y. Tang, Y. Wang, K.-M. Park, Q. Hu, J.-P. Teoh, Z. Broskova, P. Ranganathan, C. Jayakumar, microRNA program induces mammalian heart regeneration. Cell Stem Cell 15, 589–604
J. Li, H. Su, Y. Tang, G. Ramesh, I.-M. Kim, MicroRNA-150 protects the mouse heart (2014).
from ischaemic injury by regulating cell death. Cardiovasc. Res. 106, 387–397 (2015). 282. B. Icli, A. K. M. Wara, J. Moslehi, X. Sun, E. Plovie, M. Cahill, J. F. Marchini, A. Schissler,
262. Y. Li, Q. Li, O. Zhang, X. Guan, Y. Xue, S. Li, X. Zhuang, B. Zhou, G. Miao, miR-202-5p protects rat R. F. Padera, J. Shi, H.-W. Cheng, S. Raghuram, Z. Arany, R. Liao, K. Croce, C. M. Rae,
against myocardial ischemia reperfusion injury by downregulating the expression of Trpv2 M. W. Feinberg, MicroRNA-26a regulates pathological and physiological angiogenesis by
to attenuate the Ca2+ overload in cardiomyocytes. J. Cell. Biochem. 120, 13680–13693 targeting BMP/SMAD1 signaling. Circ. Res. 113, 1231–1241 (2013).
(2019). 283. T. M. Ribeiro-Rodrigues, T. L. Laundos, R. Pereira-Carvalho, D. Batista-Almeida, R. Pereira,
263. Y. Qin, Y. Yu, H. Dong, X. Bian, X. Guo, S. Dong, MicroRNA 21 inhibits left ventricular V. Coelho-Santos, A. P. Silva, R. Fernandes, M. Zuzarte, F. J. Enguita, M. C. Costa,
remodeling in the early phase of rat model with ischemia-reperfusion injury by P. Pinto-do-Ó, M. T. Pinto, P. Gouveia, L. Ferreira, J. C. Mason, P. Pereira, B. R. Kwak,
suppressing cell apoptosis. Int. J. Med. Sci. 9, 413–423 (2012). D. S. Nascimento, H. Girão, Exosomes secreted by cardiomyocytes subjected to ischaemia
264. R. A. Boon, K. Iekushi, S. Lechner, T. Seeger, A. Fischer, S. Heydt, D. Kaluza, K. Tréguer, promote cardiac angiogenesis. Cardiovasc. Res. 113, 1338–1350 (2017).
G. Carmona, A. Bonauer, A. J. G. Horrevoets, N. Didier, Z. Girmatsion, P. Biliczki, 284. Y. Peng, F. Chao, Y. Cai, W. Teng, C. Qiu, MiR-126 inhibits the proliferation of myocardial
J. R. Ehrlich, H. A. Katus, O. J. Müller, M. Potente, A. M. Zeiher, H. Hermeking, S. Dimmeler, fibroblasts by regulating EGFL7-mediated EGFR signal pathway. Int. J. Clin. Exp. Med. 10,
MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–110 (2013). 6158–6166 (2017).
265. M. S. A. Sheikh, Overexpression of miR-375 protects cardiomyocyte injury following 285. Z. Wei, S. Qiao, J. Zhao, L. Yihai, L. Qiaoling, W. Zhonghai, D. Qing, K. Lina, X. Biao,
hypoxic-reoxygenation injury. Oxid. Med. Cell. Longev. 2020, 7164069 (2020). miRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced
266. R. Li, H.-H. Geng, J. Xiao, X.-t. Qin, F. Wang, J.-h. Xing, Y.-f. Xia, Y. Mao, J.-w. Liang, X.-p. Ji, inflammatory response after myocardial ischemia-reperfusion injury. Life Sci. 232, 116632
MIR-7a/b attenuates post-myocardial infarction remodeling and protects H9c2 (2019).
cardiomyoblast against hypoxia-induced apoptosis involving Sp1 and PARP-1. Sci. Rep. 6, 286. Z. Liu, P. Ye, S. Wang, J. Wu, Y. Sun, A. Zhang, L. Ren, C. Cheng, X. Huang, K. Wang,
29082 (2016). P. Deng, C. Wu, Z. Yue, J. Xia, MicroRNA-150 protects the heart from injury by inhibiting
267. C.-L. Zhai, G.-M. Tang, G. Qian, H.-L. Hu, S.-J. Wang, D. Yin, S. Zhang, MicroRNA-98 monocyte accumulation in a mouse model of acute myocardial infarction. Circ.
attenuates cardiac ischemia-reperfusion injury through inhibiting DAPK1 expression. Cardiovasc. Genet. 8, 11–20 (2015).
IUBMB Life 71, 166–176 (2019). 287. Y. Song, C. Zhang, J. Zhang, Z. Jiao, N. Dong, G. Wang, Z. Wang, L. Wang, Localized
268. B. Bian, X.-F. Yu, G.-Q. Wang, T.-M. Teng, Role of miRNA-1 in regulating connexin 43 injection of miRNA-21-enriched extracellular vesicles effectively restores cardiac function
in ischemia–reperfusion heart injury: A rat model. Cardiovasc. Pathol. 27, 37–42 (2017). after myocardial infarction. Theranostics 9, 2346–2360 (2019).
269. L. Ge, Y. Cai, F. Ying, H. Liu, D. Zhang, Y. He, L. Pang, D. Yan, A. Xu, H. Ma, Z. Xia, 288. Z.-W. Ge, X.-L. Zhu, B.-C. Wang, J.-L. Hu, J.-J. Sun, S. Wang, X.-J. Chen, S.-P. Meng, L. Liu,
MiR-181c-5p exacerbates hypoxia/reoxygenation-induced cardiomyocyte apoptosis via Z.-Y. Cheng, MicroRNA-26b relieves inflammatory response and myocardial remodeling
targeting PTPN4. Oxid. Med. Cell. Longev. 2019, 1957920 (2019). of mice with myocardial infarction by suppression of MAPK pathway through binding
270. X. Yuan, C. Chen, L. Wang, X. He, Inhibition of microRNA-182 reduces hypoxia/re-oxygenation- to PTGS2. Int. J. Cardiol. 280, 152–159 (2019).
induced HL-1 cardiomyocyte apoptosis by targeting the nuclear respiratory factor-1/ 289. S. Lu, Y. Lu, MiR-26a inhibits myocardial cell apoptosis in rats with acute myocardial
mitochondrial transcription factor A (NRF-1/mtTFA) pathway (2017); www.ijcep.com/. infarction through GSK-3 pathway. Eur. Rev. Med. Pharmacol. Sci. 24, 2659–2666
271. D.-W. Liu, Y.-N. Zhang, H.-J. Hu, P.-Q. Zhang, W. Cui, Downregulation of microRNA-199a-5p (2020).
attenuates hypoxia/reoxygenation-induced cytotoxicity in cardiomyocytes by targeting 290. H.-Y. Wu, J.-L. Wu, Z.-L. Ni, Overexpression of microRNA-202-3p protects against
the HIF-1-GSK3-mPTP axis. Mol. Med. Rep. 19, 5335–5344 (2019). myocardial ischemia-reperfusion injury through activation of TGF-1/Smads signaling
272. Y. Wang, Y. Jiang, X. Sun, X. Shen, H. Wang, C. Dong, B. Lu, Y. Yan, Y. Lu, M. B. Fasae, B. Liu, pathway by targeting TRPM6. Cell Cycle 18, 621–637 (2019).
Y. Bai, Downregulation of miR-200a protects cardiomyocyte against apoptosis. Biomed. 291. T. Jiang, H. You, D. You, L. Zhang, M. Ding, B. Yang, A miR-1275 mimic protects
Pharmacother. 123, 109303 (2020). myocardiocyte apoptosis by regulating the Wnt/NF-B pathway in a rat model
273. Y.-S. Song, H.-W. Joo, I.-H. Park, G.-Y. Shen, Y. Lee, J. H. Shin, H. Kim, K.-S. Kim, Bone of myocardial ischemia–reperfusion-induced myocardial injury. Mol. Cell. Biochem. 466,
marrow mesenchymal stem cell-derived vascular endothelial growth factor attenuates 129–137 (2020).
cardiac apoptosis via regulation of cardiac miRNA-23a and miRNA-92a in a rat model 292. Y. Li, J. Zhou, O. Zhang, X. Wu, X. Guan, Y. Xue, S. Li, X. Zhuang, B. Zhou, G. Miao, L. Zhang,
of myocardial infarction. PLOS ONE 12, e0179972 (2017). Bone marrow mesenchymal stem cells-derived exosomal microRNA-185 represses

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 20 of 21
SCIENCE TRANSLATIONAL MEDICINE | REVIEW

ventricular remolding of mice with myocardial infarction by inhibiting SOCS2. 298. J. Dong, Z. Zhang, H. Huang, P. Mo, C. Cheng, J. Liu, W. Huang, C. Tian, C. Zhang, J. Li,
Int. Immunopharmacol. 80, 106156 (2020). MiR-10a rejuvenates aged human mesenchymal stem cells and improves heart function
293. J. Yang, Z. Fan, J. Yang, J. Ding, C. Yang, L. Chen, MicroRNA-22 attenuates myocardial after myocardial infarction through KLF4. Stem Cell Res. Ther. 9, 151 (2018).
ischemia-reperfusion injury via an anti-inflammatory mechanism in rats. Exp. Ther. Med.
12, 3249–3255 (2016).
294. H. Tan, J. Qi, B.-Y. Fan, J. Zhang, F.-F. Su, H.-T. Wang, MicroRNA-24-3p attenuates Funding: We gratefully acknowledge funding support of our cardiac research by NIH [grants
myocardial ischemia/reperfusion injury by suppressing RIPK1 expression in mice. UH3EB025765 (G.V.-N.), P41EB027062 (G.V.-N.), HL076485 (G.V.-N.), T32GM00736 (T.N.), and
Cell. Physiol. Biochem. 51, 46–62 (2018). F30HL145921 (B.L.)], NYSTEM [C32606GG (G.V.-N.)], NSF [grant ERC CELL-MET 16478 (G.V.-N.)],
295. F.-Q. Zhou, X.-F. Zhao, F.-Y. Liu, S.-S. Wang, H.-L. Hu, Y. Fang, MiR-101a attenuates and NYSTEM [grant C32606GG (G.V.-N.)]. Author contributions: B.L., B.W., X.Z., R.L., T.N., and
myocardial cell apoptosis in rats with acute myocardial infarction via targeting G.V.-N. conceptualized, outlined, and edited the manuscript. B.L., B.W., X.Z., R.L., and T.N.
TGF-/JNK signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 23, 4432–4438 surveyed relevant literature and wrote the manuscript. Competing interests: The authors
(2019). declare that they have no competing interests.
296. B.-T. Yu, N. Yu, Y. Wang, H. Zhang, K. Wan, X. Sun, C.-S. Zhang, Role of miR-133a Submitted 31 May 2020
in regulating TGF-1 signaling pathway in myocardial fibrosis after acute myocardial Accepted 19 January 2021
infarction in rats. Eur. Rev. Med. Pharmacol. Sci. 23, 8588–8597 (2019). Published 10 February 2021
297. R. Hinkel, D. Penzkofer, S. Zühlke, A. Fischer, W. Husada, Q.-F. Xu, E. Baloch, E. van Rooij, 10.1126/scitranslmed.abd0914
A. M. Zeiher, C. Kupatt, S. Dimmeler, Inhibition of microRNA-92a protects against

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


ischemia/reperfusion injury in a large-animal model. Circulation 128, 1066–1075 Citation: B. Liu, B. Wang, X. Zhang, R. Lock, T. Nash, G. Vunjak-Novakovic, Cell type–specific
(2013). microRNA therapies for myocardial infarction. Sci. Transl. Med. 13, eabd0914 (2021).

Liu et al., Sci. Transl. Med. 13, eabd0914 (2021) 10 February 2021 21 of 21
Cell type−specific microRNA therapies for myocardial infarction
Bohao Liu, Bryan Wang, Xiaokan Zhang, Roberta Lock, Trevor Nash and Gordana Vunjak-Novakovic

Sci Transl Med 13, eabd0914.


DOI: 10.1126/scitranslmed.abd0914

Downloaded from http://stm.sciencemag.org/ at George Mason University on February 11, 2021


ARTICLE TOOLS http://stm.sciencemag.org/content/13/580/eabd0914

SUPPLEMENTARY http://stm.sciencemag.org/content/suppl/2021/02/08/13.580.eabd0914.DC1
MATERIALS

RELATED http://stm.sciencemag.org/content/scitransmed/12/573/eabb3336.full
CONTENT
http://stm.sciencemag.org/content/scitransmed/12/561/eaay1318.full
http://stm.sciencemag.org/content/scitransmed/11/493/eaau1168.full
http://stm.sciencemag.org/content/scitransmed/7/279/279ra38.full
REFERENCES This article cites 295 articles, 36 of which you can access for free
http://stm.sciencemag.org/content/13/580/eabd0914#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science Translational Medicine (ISSN 1946-6242) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. The title Science Translational Medicine is a
registered trademark of AAAS.
Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement
of Science. No claim to original U.S. Government Works

You might also like