Open Access

MicroRNA‑378: An important player in cardiovascular diseases (Review)

  • Authors:
    • Huan Wang
    • Jingjing Shi
    • Jiuchong Wang
    • Yuanhui Hu
  • View Affiliations

  • Published online on: July 25, 2023     https://doi.org/10.3892/mmr.2023.13059
  • Article Number: 172
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cardiovascular disease (CVD) is a common chronic clinical condition and is the main cause of death in humans worldwide. Understanding the genetic and molecular mechanisms involved in the development of CVD is essential to develop effective prevention strategies and therapeutic measures. An increasing number of CVD‑related genetic studies have been conducted, including those on the potential roles of microRNAs (miRs). These studies have demonstrated that miR‑378 is involved in the pathological processes of CVD, including those of myocardial infarction, heart failure and coronary heart disease. Despite the potential importance of miR‑378 CVD, a comprehensive summary of the related literature is lacking. Thus, the present review aimed to summarize the findings of previous studies on the roles and mechanisms of miR‑378 in a variety of CVDs and provide an up‑to date basis for further r research targeting the prevention and treatment of CVDs.

Introduction

Cardiovascular disease (CVD) is, as a common chronic clinical disease characterized by high incidence, high disability and high mortality rates. The number of CVD patients in China is ~290 million and the incidence and mortality rates continue to increase worldwide (1). Treatment of CVD has become a subject of interest in modern medicine. Significant progress has been made in the treatment of CVD, the clinical application of new drugs and continuous improvements in medical advances, however, CVD still exacts a heavy toll on the health of patients, causes undue economic pressure and remains the commonest cause of mortality (2). Thus, it is necessary to understand the potential molecular mechanisms that regulate the progress of CVD to develop highly effective prevention strategies and therapeutic measures.

In the early stages of heart failure, the heart compensates for abnormal hemodynamics by promoting cardiomyocyte hypertrophy and inducing abnormal expression of genes (3,4). However, long-term myocardial hypertrophy leads to ventricular remodeling in which myocardial fibroblasts promote cardiomyocyte hypertrophy and extracellular matrix deposition through autocrine and paracrine mechanisms, leading to further reduction in cardiac function (5). MicroRNAs (miRNAs/miRs) are noncoding RNAs (ncRNAs) containing 19–22 nucleotides that exist stably in the blood and regulate posttranscriptional gene expression by binding to target mRNAs and reducing protein synthesis (6). miRNAs serve important regulatory roles in biological processes such as cell proliferation, apoptosis and differentiation (7) and are involved in the pathogenesis of numerous diseases, such as ovarian cancer, glioma, oral squamous cell carcinoma, gastric cancer and hepatocellular carcinoma (710). Abnormal miRNA expression is involved in the development and occurrence of CVDs (11). miRNAs may serve as a new mode of cell-to-cell communication via the regulation of gene expression in target cells and tissues and play a core role in gene regulation (12). The characteristics of miRNAs are similar in males and females and in individuals of different ages; thus, miRNAs represent potential indicators of different diseases (13).

miR-378 (also known as miR-378a-3p) is a cardioprotective miRNA involved in the regulation of the occurrence and development of a variety of heart diseases (14,15). Several variants of miR-378 exist, including miR-378a/b/c/d/e/f/g/h/i/j, all of which are encoded by different genomic sites but possess common regulatory targets because they share the same seed sequence.

miR-378 is involved in the pathological processes of heart diseases, including myocardial infarction, heart failure and coronary heart disease (CHD). The pathological processes involve myocardial hypertrophy, myocardial cell fibrosis, myocardial remodeling, mitochondrial energy metabolism and angiogenesis (16,17).

Notably, the physiological expression of miR-378 is associated with the growth and development of individuals. For example, the expression of miR-378 gradually increases with age in mouse hearts, as shown in measurements of 16-day-old mouse fetuses and 1, 2, 4 week- and 10 month-old mice (18). This trend of increasing expression was confirmed in a second study of mouse fetuses examined at 10 and 14 days (19). Thus, the expression of miR-378 in the heart gradually increases from the fetal stage to adulthood following significant growth of the heart and continues to increase even after heart development is complete after birth.

There are no reviews involved in the function of miR-378 in CVD. Therefore, the present review summarized the current understanding of the roles and mechanisms of action of miR-378 in CVD. The aim is to provide a comprehensive review of the current state of research in support of more in-depth scientific and clinical studies.

miR-378 regulation in heart disease

Diabetic cardiomyopathy (DCM)

DCM is a special form of CVD caused by resistance to insulin metabolism in heart tissue, compensatory hyperinsulinemia and hyperglycemia. The occurrence of DCM is independent of other cardiac risk factors (20) and an increasing number of studies have shown that oxidative stress, inflammation, mitochondrial dysfunction, renin-angiotensin system activation and myocardial cell apoptosis are involved in the pathogenesis of DCM (2124).

In patients with diabetes, high blood glucose levels can cause an imbalance in the oxidative stress state, affecting cell apoptosis, hypertrophy, fibrosis, calcium homeostasis and contractile function, leading to DCM. However, even if blood glucose levels become relatively stable, hyperglycemia-induced myocardial damage cannot be prevented. This phenomenon is called hyperglycemia or metabolic memory (25,26). A meta-analysis found that the risk of heart failure in patients with diabetes cannot be avoided even after the effective control of coronary artery disease or hypertension (27). Costantino et al (28) observed that 48 miRNAs returned to normal expression levels following 3 weeks of intensive insulin treatment in diabetic mice; however, the expression of 268 miRNAs did not return to their original levels. Abnormally expressed miRNAs are involved in pathways that mediate cardiomyocyte apoptosis, fibrosis, hypertrophy, autophagy, oxidative stress and heart failure (29). miR-378 is an overexpressed miRNA that cannot be restored to its original expression level and thus is a key promoter of apoptosis (3032). As miR-378 is involved in diabetic myocardial damage, it may serve as a therapeutic target for the prevention and treatment of DCM.

Durr et al (33) demonstrated that diabetes mellitus is associated with increased mitochondrial (MT) miR-378a level, which, in turn, is linked to a decrease in MT-ATP6 abundance, a protein encoded by the mitochondrial genome. Furthermore, these authors demonstrated the potential of miR-378a to bind and downregulate the expression of MT-ATP6, leading to a decrease in the function of ATP synthase. Treatment with miR-378a-antagomir improved cardiac systolic function in mice treated with streptozotocin. In addition, in a genetic mouse model of miR-378a deletion, improvement in cardiac dysfunction in patients with type 2 diabetes was observed, kcnq10t1 may interact with miR378a, affecting ATP synthase activity by regulating the expression of MT-ATP6 (33).

Myocardial damage caused by doxorubicin (DOX)

DOX is a highly effective anticancer drug that is widely used in cancer treatment; however, severe cardiotoxicity greatly limits its clinical application (34). More research attention is being paid to miRNAs and their role in DOX-induced cardiotoxicity at the genetic levels, with improved prospects emerging (35,36).

The mechanism of DOX-induced cell damage is probably related to cell apoptosis and endoplasmic reticulum stress (ERS) can trigger an important signaling pathway leading to cell apoptosis (37). ERS generally affects the function of myocardial mitochondria, leading to mitochondrial dysfunction and energy imbalance in myocardial cells, which affect heart function and eventually lead to acute or chronic heart failure (38,39).

The miRNA profile of heart tissue in DOX-induced cardiotoxic mice or rat models was characterized after 2–8 weeks of treatment (40,41). Chen et al (42) observed changes in the miRNA profile of DOX-treated H9C2 cells within 24 h and identified 67 miRNAs with downregulated expression. The genes encoding theses miRNAs were mainly involved in extracellular matrix receptor interaction, focal adhesion and the PI3K-Akt signaling pathway. miR-378-3p and −5p were among the miRNAs exhibiting downregulated expression.

Wang et al (43) explained the mechanism of DOX-induced cardiotoxicity as occurring via the effect of miR-378 on mitochondrial energy metabolism and ERS. miR-378 mainly targets LDHA, reducing lactate dehydrogenase content and improving mitochondrial energy metabolism. LDHA can also target PPIA to regulate cell apoptosis; thus, miR-378 participates in the regulation of energy metabolism and cell apoptosis by affecting LDHA and PPIA (43). miR-378 is also involved in ERS-mediated apoptosis through its regulation of the expression of the calcium-binding endoplasmic reticulum protein calumenin (44). Calumenin serves important roles via two mechanisms: i) inhibiting the levels of GRP78, p-PERK and p-eIF2a, which reduces ERS-mediated apoptosis and eases cell apoptosis; and ii) inhibiting the activities of SERCA2 and the calcium release channel RyR2 to maintain a steady state of calcium circulation in cardiomyocytes, which then maintains the contractile and diastolic functions of cardiomyocytes (45).

Coronary heart disease
Improving hypoxia

Acute myocardial infarction (AMI) is a common type of CHD and caused by acute ischemia and hypoxia (Table I and Fig. 1). Hypoxia-inducible factor (HIF) is the main regulator of cellular responses to hypoxia (46). Under hypoxic conditions, HIF-α is activated and transferred from the cytoplasm to the nucleus, thereby activating the hypoxia adaptation pathway, which reduces oxygen consumption and improves the cell survival rate by promoting angiogenesis, erythropoiesis and iron metabolism (47,48). Continuous hypoxia eventually leads to cell apoptosis and even necrosis (49).

Figure 1.

miRNA-378 is involved in mediating myocardial hypoxia, angiogenesis, cell apoptosis, autophagy, hypertrophy and fibrosis. miRNA/miR, microRNA.

Table I.

miR-378 roles and changes in coronary heart disease.

Table I.

miR-378 roles and changes in coronary heart disease.

First author/s, year Effect Types of miR-378 Trend Model Condition Pathway/targets (Refs.)
Krown et al, 1996; Chang and Karin, 2001 Improving hypoxia miR-378-3p Upregulated H9C2 cells and exosomes Hypoxia HIF-1, TNF, MAPK, mTOR (56,57)
Xing et al, 2014 Promoting angiogenesis miR-378a-5p Upregulated MSCs Hypoxia SuFu and Fus-1 (65)
Lee et al, 2007 miR-378 Upregulated CD34+ progenitor cells STEMI patients (67)
Zhou et al, 2021 Improving cell apoptosis miR-378 Downregulated MI/RI model in mice Ligating the LAD MAPK1 (71)
Zhang et al, 2020 Improving autophagy miR-378-3p Downregulated Peripheral blood Patients with UA and patients with ischemic cardiomyopathy/MI lncRNA NEAT1/miR-378-3p/Atg12 (72)
MI/RI injury in mice Ligating the distal part of the left ECA
Myocyte isolated from the heart of the newborn rat Hypoxia
Li et al, 2019 Improved prognosis miR-378 Downregulated Plasma Patients who underwent PCI with DES implantation - (74)
Shen et al, 2021 Downregulated ULMCAD patients who underwent CABG (75)
Kwee et al, 2019 Worse prognosis miR-378a-3p Downregulated Plasma NSTE-ACS - (78)

[i] miR, microRNA; MSCs, mesenchymal stem cells; MI/RI, myocardial ischemia reperfusion; ECA, external carotid artery; LAD, left anterior descending; UA, unstable angina; STEMI, ST elevated myocardial infarction; MI, myocardial infarction; PCI, percutaneous coronary intervention; DES, drug-eluting stent; ULMCAD, unprotected left main coronary artery disease; CABG, coronary artery bypass graft; NSTEMI, non-ST elevated myocardial infarction; ACS, acute coronary syndrome; HIF, hypoxia-inducible factor; lncRNA, long non-coding RNA; MAPK1, mitogen-activated protein kinase 1.

Previous studies have confirmed that hypoxia, called ‘hypoxemia’, can regulate the expression of miRNAs involved in the HIF signaling pathway (5052). Exosomes are signaling mediators that promote cell-to-cell communication by transporting cytoplasmic components, including miRNAs, mRNAs and proteins (53). Under hypoxic conditions, miRNAs in cells and exosomes undergo certain changes and miR-378 is considered a potential biomarker of cell hypoxia (54). In vitro experiments have confirmed that exosomes under early hypoxic conditions can reduce hypoxia-induced apoptosis by overexpressing cardioprotective miRNAs, such as miR-378-3p (55). Hypoxia also alters the expression levels of 92 miRNAs in H9C2 cells and 62 miRNAs in exosomes. Differentially expressed exosomal miRNAs mainly participate in pathways related to cell viability, including the HIF-1, TNF, MAPK and mTOR signaling pathways (56,57).

Promoting angiogenesis

Angiogenesis is a key and complex process in the treatment of CVD and is determined by the interaction between proangiogenic and antiangiogenic factors (58,59). During the pathological process of CHD, there is an imbalance between oxygen consumption and supply in cardiomyocytes, which can damage vascular function. Therefore, angiogenesis is considered to be the primary cause of CHD (60). Thus, promoting angiogenesis and improving myocardial tissue perfusion and the recovery of myocardial function are the primary aims in treating CHD (61,62).

miRNAs have certain advantages in regulating angiogenic factors to improve vascular function in patients with CHD (63). Among these, a class of ncRNAs, such as miR-126 and miR-378, can regulate the expression of anti-angiogenic genes and promote angiogenesis. These ncRNAs can improve angiogenesis in endothelial cells and reduce the proliferation and migration of ectopic vascular smooth muscle cells, reducing the risk of CHD (64).

Xing et al (65) confirmed that the overexpression of miR-378a-5p in mesenchymal stem cells under hypoxic conditions inhibit mesenchymal stem cell apoptosis and promote the expression of angiogenesis-related genes (VEGFA, PDGF-β and TGF-β1). The expression of CD34+ progenitor cells is significantly increased in patients with ST elevated myocardial infarction and exhibit high proangiogenic activity, which is closely related to the release of paracrine factors (66), including the upregulation of miR-378 expression. miR-378 may promote angiogenesis by targeting the expression of suppressor of fused (SuFu) and Fus-1 (67). The promotion of angiogenesis by upregulating CD34 and miR-378 expression is a unique repair mechanism in patients with AMI (68).

Improving cell apoptosis

Myocardial ischemia/reperfusion resulting in cardiomyocyte apoptosis and necrosis is the main feature of AMI (69,70). This process causes a decrease in cardiomyocyte function and ventricular remodeling, eventually leading to heart failure.

A mouse model of myocardial ischemia-reperfusion injury (MI/RI) was established by ligating the left anterior descending branch of the coronary artery. The level of miR-378 in the MI/RI model mice decreased, while that of MAPK1 increased, whereas after intervention with isoproterenol (ISO), miR-378 and MAPK1 were upregulated and downregulated, respectively. This confirmed that ISO improved cardiomyocyte apoptosis by regulating miR-378 and MAPK1 in the MI/RI model (71).

Improving autophagy

Long non-coding RNAs (lncRNAs) interact with miRNAs as competitive endogenous RNAs (ceRNAs) and participate in regulating target gene expression. Zhang et al (72) confirmed that lncRNA NEAT1 is highly expressed in the peripheral blood of patients with myocardial infarction and in mouse cardiomyocytes. lncRNA NEAT1 binds to and inhibits the expression of miR-378a-3p, thereby promoting the proliferation and migration of cardiomyocytes. Inhibiting the expression of miR-378a-3p facilitates the expression of Atg12 and related autophagy factors, thereby enhancing the autophagy of cardiomyocytes. Therefore, the proliferation of cardiomyocytes in patients with myocardial infarction can be modulated by inhibiting the NEAT1/miR-378-3p/Atg12 axis.

Prognosis

miR-378 is considered as a marker of CHD risk and of the degree of coronary stenosis and restenosis following coronary stent intervention (64,73). The plasma level of miR-378 expression in patients with CHD is significantly downregulated and negatively correlated with the Gensini score (74). Patients with unprotected left main coronary artery disease were followed-up in the first, second and third years after coronary angiography to observe the incidence of major adverse cardiac and cerebrovascular event (MACCE) and to detect the levels of circulating miR-378 in the peripheral blood. The results showed that a high level of miR-378 expression can independently predict a low incidence of MACCE (75). The expression of osteopontin (OPN), a matrix cell protein that regulates inflammation and biomineralization, also increases under a number of pathological conditions (76). Increased OPN levels can effectively predict the mortality rate in patients with heart failure (77). Kwee et al (78) measured the expression of miRNAs in 186 patients in the TRILOGY-ACS trial and discovered that the expression of miR-378a-3p was closely associated with OPN levels, which are linked with cardiovascular death, myocardial infarction and stroke.

Heart failure

In patients with early heart failure, the level of miR-378 expression was relatively upregulated and gradually decreased with the aggravation of heart failure, whereas the level of NT-proBNP expression showed an upward trend with heart failure progression (Table II and Fig. 1). This may be related to the compensatory function of the heart. Therefore, it is hypothesized that the upregulation of miR-378 expression is a self-compensatory effect of the heart in the early stages of heart failure (79). In patients with heart failure, evaluation of the expression levels of circulating miRNAs before and after acute exhaustive exercise showed that the level of miR-378 was elevated and was not significantly related to exercise endurance, muscle damage, or inflammation (80). However, with the progression of heart failure, a series of pathophysiological mechanisms, such as cardiomyocyte hypertrophy and myocardial fibrosis, cause ventricular remodeling, which further aggravates heart failure and the miR-378 expression levels vary accordingly (80).

Table II.

miR-378 roles and changes in heart failure.

Table II.

miR-378 roles and changes in heart failure.

First author/s, year Effect Types of miR-378 Trend Model Intervention method Target genes/pathways (Refs.)
Bernardo et al, 2010; Ganesan et al, 2013 Cardiomyocyte hypertrophy miR-378 Downregulated Isolated neonatal rat cardio-myocytes or cardiac fibroblasts Phenylephrine MAPK1, KSR1, GRB2 and IGF1R (84,85)
Downregulated Left ventricular myocardium Transgenic overexpression of the β1-adrenergic receptor
Downregulated Left ventricular myocardium Not discussed
Barut et al, 2021 Cardiomyocyte hypertrophy miR-378a-3p Upregulated Left ventricular hypertrophy Not discussed Not discussed (87)
Moghiman et al, 2021 Myocardial fibrosis miR-378 Upregulated Neonatal CMs and CFs miR-378 mimics MKK6/p38MAPK/Smad2/3 (94)
Upregulated TAC Intravenous injections of chemically modified miR-378 mimics
Sun et al, 2019 Downregulated Male C57BL/6 mice Ligating the LAD LncRNA PCFL/miR-378/GRB2 (95)
Downregulated CFs Transfection of lncRNA PCFL plasmids

[i] miR, microRNA; CMs, cardiomyocytes; CFs, cardiac fibroblasts; TAC, transverse aortic constriction; LAD, left anterior descending; lncRNA, long non-coding RNA; KSR1, kinase suppressor of ras 1; GRB2, growth factor receptor-bound protein 2; MAPK1, mitogen-activated protein kinase 1; IGF1R; insulin-like growth factor1 receptor; MKK6, mitogen-activated protein kinase kinase 6.

Adenosine triphosphate sensitive potassium channels (KATP) serve as gating channels connecting vascular reactivity, metabolism and ischemic protection and are potential targets for the treatment of heart failure (81). A causal relationship exists between lower serum apolipoprotein A-I (ApoA-I) concentrations and the risk of heart failure. In one study, a total of 634 patients were retrospectively selected, including 317 patients with lo ApoA-I concentrations (<120 mg/dl) and 317 patients with high ApoA-I concentrations (≥120 mg/dl). Identification of extracellularly derived miRNA expression profiles through next-generation sequencing revealed that KATP rs41294036 was related to an increased risk of lower ApoA-I levels and an increased incidence rate of heart failure, particularly heart failure with preserved ejection fraction. Furthermore, patients carrying the rs141294036 CC genotype were associated with an increased risk of heart failure-related rehospitalization (82).

Cardiomyocyte hypertrophy (CH)

CH is an adaptive response of the heart to pressure or volume overload to maintain cardiac function during the early stages (83). However, persistent cardiac hypertrophy often leads to maladaptive cardiac remodeling and increases the risk of heart failure and sudden death (83).

CH is primarily involved in physiological and pathological hypertrophy. However, no association has been observed between physiological hypertrophy and heart failure. Physiological hypertrophy mainly occurs during postnatal development, exercise response and pregnancy and is an adaptive response that can protect the heart (84). CH is reversible and the main pathological process involves the activation of the IGF1-PI3K (p110a)-Akt pathway. Pathological hypertrophy mainly occurs during the development of heart diseases (hypertension, myocardial infarction and heart failure), manifesting as an accumulation of extracellular matrix, death or apoptosis of cardiomyocytes and an imbalance in calcium homeostasis, leading to a decrease in contraction and diastole of cardiomyocytes. This change is irreversible and is usually related to increases in various endogenous factors (such as angiotensin II, endothelin 1 and norepinephrine) and affects cardiomyocyte hypertrophy via the RAS signaling pathway (84).

miR-378 is an endogenous protective modulator of the heart. Overexpression of miR-378 not only reduces cardiomyocyte hypertrophy during the stress-loaded heart adaptation period but also reduces cardiac hypertrophy by targeting Ras signaling pathways, such as growth factor receptor-binding protein 2 (GRB2), kinase inhibitor Ras 1 (KSR1) and mitogen-activated protein kinase (MAPK)1. miR-378 target genes are enriched in MAPK pathway, including genes encoding MAPK1, insulin-like growth factor receptor 1, GRB2 and KSR1 (85).

Chen et al (86) systematically studied the ceRNA regulatory network related to the circRNAs involved in CH. They used a public database of online prediction websites to predict, and screen for, differentially expressed mRNAs and miRNAs, ultimately identifying circRNAs related to CH. To the best of the authors' knowledge, this is the first study to establish a ceRNA regulatory network associated with myocardial hypertrophy. A total of three mRNAs, four miRNAs (including miR-378a-3p) and four related circRNAs may play key roles in CH. Barut et al (87) hypothesized that high circulating levels of miRNA-378 could serve as a new biomarker for the diagnosis of left ventricular hypertrophy (LVH).

Myocardial fibrosis

Excessive myocardial fibrosis is the main pathological process underlying heart remodeling and failure (88,89). Myocardial fibrosis is characterized by the accumulation of fibroblasts, excessive deposition of extracellular matrix proteins and increased levels of collagen and matrix metalloproteinases (MMPs) (90,91). Abnormal expression of numerous miRNAs during excessive hemodynamic pressure is a key component of heart remodeling (92,93).

miR-378 can reduce the expression of collagen and MMP9 by inhibiting the p38MAPK and Smad2/3 signaling pathways, thereby inhibiting myocardial fibrosis. miR-378 mimics reduced myocardial fibrosis in rats with transverse aortic constriction (TAC) (94). A possible mechanism involves miR-378 inhibition of the p38MAPK and Smad2/3 signaling pathways by directly targeting MAPK6 in an extracellular vesicle-dependent manner. Therefore, the expression of collagen and MMP in cardiac fibroblasts cells (CFs) is regulated and that in myocardial fibrosis is inhibited (79). miR-378 also exerts an anti-myocardial fibrotic effect through a ceRNA mechanism. The combination of the pro-cardiac fibrotic lncRNA and miR-378 relieves the inhibition of GRB2, resulting in elevated expression of TGF-β1, which promotes myocardial fibrosis (95). In addition, heat shock transcription factor 1 (HSF1) is a protective factor in the heart and the level of miR-378 expression in TAC mice with HSF1 knock out was significantly reduced, resulting in increased myocardial fibrosis (96).

Aortic stenosis

Aortic stenosis is a common heart valve disease that accounts for 43% of adult patients with valvular heart diseases (97). The most common complication is LVH, which is closely related to the occurrence of cardiovascular events in patients with aortic stenosis (AS) and is an independent predictor of an adverse prognosis for these patients (98101). miR-378 mediates the pathological process of LVH through the MAPK signaling pathway (85,102). Plasma miR-378 levels in patients with AS were significantly reduced and negatively correlated with left ventricular mass index. The plasma level of miR-378 is an independent predictor of LVH in patients with AS and to a certain extent reflects heart remodeling caused by pressure overload (103).

Conclusions

As a cardioprotective miRNA, miR-378 regulates the development of various CVDs. In doing so, miR-378 participates in numerous processes, including myocardial hypertrophy, myocardial cell fibrosis, myocardial remodeling, mitochondrial energy metabolism and angiogenesis, among other processes waiting to be discovered. For example, Fig. 1 shows that miRNA-378 plays a significant role in the pathogenesis of CHD and heart failure. The main manifestation is that in terms of CHD, miRNA-378 is involved in mediating myocardial cell hypoxia, promoting angiogenesis, promoting cell apoptosis and autophagy and has certain value for the prognosis of CHD; In terms of heart failure, miRNA-378 is involved in mediating myocardial cell hypertrophy and myocardial fibrosis levels. Thus, regulating the level of miRNA-378 has certain value in the treatment of CHD and heart failure and miR-378 may be a vital regulator of CVDs.

Acknowledgements

Not applicable.

Funding

Funding was provided by the National Natural Science Foundation of China (grant no. 8207449).

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Authors' contributions

HW retrieved literature and drafted the manuscript. JS and JW read and classified the literature based on the different types of diseases. YH supervised the study and offered advice. HW, JS, JW and YH reviewed and classified the literature based on the different types of diseases, and wrote and revised the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AMI

acute myocardial infarction

AS

aortic stenosis

CHD

coronary heart disease

CVD

cardiovascular disease

DCM

diabetic cardiomyopathy

DOX

doxorubicin

ER

endoplasmic reticulum

HIF

hypoxia-inducible factor

KATP

adenosine triphosphate-sensitive potassium channels

lncRNA

long non-coding RNA

LVH

left ventricular hypertrophy

miRNA/miR

microRNA

MMPs

matrix metalloproteinases

ncRNA

non-coding RNA

OPN

osteopontin

PPIA

peptidylprolyl isomerase A

GRB2

growth factor receptor-binding protein 2

KSR1

kinase inhibitor Ras 1

MAPK1

mitogen-activated protein kinase 1

HSF1

heat shock transcription factor 1

References

1 

Triposkiadis F, Xanthopoulos A and Butler J: Cardiovascular aging and heart failure: JACC review topic of the week. J Am Coll Cardiol. 74:804–813. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, et al: Heart disease and stroke statistics-2017 update: A report from the American heart association. Circulation. 135:e146–e603. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Hennersdorf MG and Strauer BE: Arterial hypertension and cardiac arrhythmias. J Hypertens. 19:167–177. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Vakili BA, Okin PM and Devereux RB: Prognostic implications of left ventricular hypertrophy. Am Heart J. 141:334–341. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Aimo A, Panichella G, Barison A, Maffei S, Cameli M, Coiro S, D'Ascenzi F, Di Mario C, Liga R, Marcucci R, et al: Sex-related differences in ventricular remodeling after myocardial infarction. Int J Cardiol. 339:62–69. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Cheng M, Yang J, Zhao X, Zhang E, Zeng Q, Yu Y, Yang L, Wu B, Yi G, Mao X, et al: Circulating myocardial microRNAs from infarcted hearts are carried in exosomes and mobilise bone marrow progenitor cells. Nat Commun. 10:9592019. View Article : Google Scholar : PubMed/NCBI

7 

Zhou Y, Xia Z, Cheng Z, Xu G, Yang X, Liu S and Zhu Y: Inducible microRNA-590-5p inhibits host antiviral response by targeting the soluble interleukin-6 (IL6) receptor. J Biol Chem. 293:18168–18179. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Zhang L, Wang YH and Wang L: MiRNA-8073 targets ZnT1 to inhibit malignant progression of ovarian cancer. Eur Rev Med Pharmacol Sci. 23:6062–6069. 2019.PubMed/NCBI

9 

Mei JW, Yang ZY, Xiang HG, Bao R, Ye YY, Ren T, Wang XF and Shu YJ: MicroRNA-1275 inhibits cell migration and invasion in gastric cancer by regulating vimentin and E-cadherin via JAZF1. BMC Cancer. 19:7402019. View Article : Google Scholar : PubMed/NCBI

10 

Zhao B, Lu Y, Cao X, Zhu W, Kong L, Ji H, Zhang F, Lin X, Guan Q, Ou K, et al: MiRNA-124 inhibits the proliferation, migration and invasion of cancer cell in hepatocellular carcinoma by downregulating lncRNA-UCA1. Onco Targets Ther. 12:4509–4516. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Nagy O, Barath S and Ujfalusi A: The role of microRNAs in congenital heart disease. EJIFCC. 30:165–178. 2019.PubMed/NCBI

12 

Fisher JN, Terao M, Fratelli M, Kurosaki M, Paroni G, Zanetti A, Gianni M, Bolis M, Lupi M, Tsykin A, et al: MicroRNA networks regulated by all-trans retinoic acid and Lapatinib control the growth, survival and motility of breast cancer cells. Oncotarget. 6:13176–13200. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Taylor DD and Gercel-Taylor C: MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 110:13–21. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Huang N, Wang J, Xie W, Lyu Q, Wu J, He J, Qiu W, Xu N and Zhang Y: MiR-378a-3p enhances adipogenesis by targeting mitogen-activated protein kinase 1. Biochem Biophys Res Commun. 457:37–42. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Ikeda K, Horieinoue K, Ueno T, Suzuki T, Sato W, Shigekawa T, Osaki A, Saeki T, Berezikov E, Mano H and Inoue S: miR-378a-3p modulates tamoxifen sensitivity in breast cancer MCF-7 cells through targeting GOLT1A. Sci Rep. 5:131702015. View Article : Google Scholar : PubMed/NCBI

16 

Chen LT, Xu SD, Xu H, Zhang JF, Ning JF and Wang SF: MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Med Oncol. 29:1673–1680. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Welten SM, Goossens EA, Quax PH and Nossent AY: The multifactorial nature of microRNAs in vascular remodelling. Cardiovasc Res. 110:6–22. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Knezevic I, Patel A, Sundaresan NR, Gupta MP, Solaro RJ, Nagalingam RS and Gupta M: A novel cardiomyocyte-enriched microRNA, miR-378, targets insulin-like growth factor 1 receptor: Implications in postnatal cardiac remodeling and cell survival. J Biol Chem. 287:12913–12926. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Porrello ER, Johnson BA, Aurora AB, Simpson E, Nam YJ, Matkovich SJ, Dorn GW II, van Rooij E and Olson EN: MiR-15 family regulates postnatal mitotic arrest of cardiomyocytes. Circ Res. 109:670–679. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Jia G, Whaley-Connell A and Sowers JR: Diabetic cardiomyopathy: A hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. 61:21–28. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Bugger H and Abel ED: Molecular mechanisms of diabetic cardiomyopathy. Diabetologia. 57:660–671. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Zhou X, Tao Y, Chen Y, Xu W, Qian Z and Lu X: Serum chemerin as a novel prognostic indicator in chronic heart failure. J Am Heart Assoc. 8:e0120912019. View Article : Google Scholar : PubMed/NCBI

23 

Mages C, Gampp H, Syren P, Rahm AK, André F, Frey N, Lugenbiel P and Thomas D: Electrical ventricular remodeling in dilated cardiomyopathy. Cells. 10:27672021. View Article : Google Scholar : PubMed/NCBI

24 

Mohananey D, Mewhort H, Shekhar S, Mohananey A, Chaudhary R, Gaglianello N and Ramakrishna H: Heart failure trial update-analysis of recent data. J Cardiothorac Vasc Anesth. 35:2792–2800. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Ceriello A: Hypothesis: The ‘metabolic memory’, the new challenge of diabetes. Diabetes Res Clin Pract. 86 (Suppl 1):S2–S6. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Paneni F, Volpe M, Lüscher TF and Cosentino F: SIRT1, p66(Shc), and Set7/9 in vascular hyperglycemic memory: Bringing all the strands together. Diabetes. 62:1800–1807. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Castagno D, Baird-Gunning J, Jhund PS, Biondi-Zoccai G, MacDonald MR, Petrie MC, Gaita F and McMurray JJ: Intensive glycemic control has no impact on the risk of heart failure in type 2 diabetic patients: Evidence from a 37,229 patient meta-analysis. Am Heart J. 162:938–948.e2. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Costantino S, Paneni F, Lüscher TF and Cosentino F: MicroRNA profiling unveils hyperglycaemic memory in the diabetic heart. Eur Heart J. 37:572–576. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Maher TM, Corte TJ, Fischer A, Kreuter M, Lederer DJ, Molina-Molina M, Axmann J, Kirchgaessler KU, Samara K, Gilberg F and Cottin V: Pirfenidone in patients with unclassifiable progressive fibrosing interstitial lung disease: A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Resp Med. 8:147–157. 2020. View Article : Google Scholar

30 

Yang BF, Lu YJ and Wang ZG: MicroRNAs and apoptosis: Implications in the molecular therapy of human disease. Clin Exp Pharmacol Physiol. 36:951–960. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Tijsen AJ, van der Made I, van den Hoogenhof MM, Wijnen WJ, van Deel ED, de Groot NE, Alekseev S, Fluiter K, Schroen B, Goumans MJ, et al: The microRNA-15 family inhibits the TGFβ-pathway in the heart. Cardiovasc Res. 104:61–71. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Li R, Geng HH, Xiao J, Qin XT, Wang F, Xing JH, Xia YF, Mao Y, Liang JW and Ji XP: miR-7a/b attenuates post-myocardial infarction remodeling and protects H9c2 cardiomyoblast against hypoxia-induced apoptosis involving Sp1 and PARP-1. Sci Rep. 6:290822016. View Article : Google Scholar : PubMed/NCBI

33 

Durr AJ, Hathaway QA, Kunovac A, Taylor AD, Pinti MV, Rizwan S, Shepherd DL, Cook CC, Fink GK and Hollander JM: Manipulation of the miR-378a/mt-ATP6 regulatory axis rescues ATP synthase in the diabetic heart and offers a novel role for lncRNA Kcnq1ot1. Am J Physiol Cell Physiol. 322:C482–C495. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Kalyanaraman B: Teaching the basics of the mechanism of doxorubicin-induced cardiotoxicity: Have we been barking up the wrong tree? Redox Biol. 29:1013942020. View Article : Google Scholar : PubMed/NCBI

35 

Xu T, Liu N, Shao Y, Huang Y and Zhu D: MiR-218 regulated cardiomyocyte differentiation and migration in mouse embryonic stem cells by targeting PDGFRα. J Cell Biochem. 120:4355–4365. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Tong Z, Jiang B, Wu Y, Liu Y, Li Y, Gao M, Jiang Y, Lv Q and Xiao X: MiR-21 protected cardiomyocytes against doxorubicin-induced apoptosis by targeting BTG2. Int J Mol Sci. 16:14511–14525. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Xin W, Li X, Lu X, Niu K and Cai J: Involvement of endoplasmic reticulum stress-associated apoptosis in a heart failure model induced by chronic myocardial ischemia. Int J Mol Med. 27:503–509. 2011.PubMed/NCBI

38 

Yang Y, Zhang H, Li X, Yang T and Jiang Q: Effects of PPARα/PGC-1α on the energy metabolism remodeling and apoptosis in the doxorubicin induced mice cardiomyocytes in vitro. Int J Clin Exp Pathol. 8:12216–12224. 2015.PubMed/NCBI

39 

Cappetta D, Rossi F, Piegari E, Quaini F, Berrino L, Urbanek K and De Angelis A: Doxorubicin targets multiple players: A new view of an old problem. Pharmacol Res. 127:4–14. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Zhu JN, Fu YH, Hu ZQ, Li WY, Tang CM, Fei HW, Yang H, Lin QX, Gou DM, Wu SL and Shan ZX: Activation of miR-34a-5p/Sirt1/p66shc pathway contributes to doxorubicin-induced cardiotoxicity. Sci Rep. 7:118792017. View Article : Google Scholar : PubMed/NCBI

41 

Zhao L, Qi Y, Xu L, Tao X, Han X, Yin L and Peng J: MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox Biol. 15:284–296. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Chen Y, Xu Y, Deng Z, Wang Y, Zheng Y, Jiang W and Jiang L: MicroRNA expression profiling involved in doxorubicin-induced cardiotoxicity using high-throughput deep-sequencing analysis. Oncol Lett. 22:5602021. View Article : Google Scholar : PubMed/NCBI

43 

Wang Y, Zhang Q, Wei C, Zhao L, Guo X, Cui X, Shao L, Long J, Gu J and Zhao M: MiR-378 modulates energy imbalance and apoptosis of mitochondria induced by doxorubicin. Am J Transl Res. 10:3600–3609. 2018.PubMed/NCBI

44 

Wang Y, Cui X, Wang Y, Fu Y, Guo X, Long J, Wei C and Zhao M: Protective effect of miR378* on doxorubicin-induced cardiomyocyte injury via calumenin. J Cell Physiol. 233:6344–6351. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Sahoo SK and Kim DH: Characterization of calumenin in mouse heart. BMB Rep. 43:158–163. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Jiang BH, Rue E, Wang GL and Semenza GL: Dimerization, DNA binding, and transactivation properties of hypoxia-inducible factor 1. J Biol Chem. 271:17771–17778. 1996. View Article : Google Scholar : PubMed/NCBI

47 

Semenza GL: Hypoxia-inducible factors in physiology and medicine. Cell. 148:399–408. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Nallamshetty S, Chan SY and Loscalzo J: Hypoxia: A master regulator of microRNA biogenesis and activity. Free Radic Biol Med. 64:20–30. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Greijer AE and van de Wall E: The role of hypoxia inducible factor 1 (HIF-1) in hypoxia induced apoptosis. J Clin Pathol. 57:1009–1014. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Azzouzi HE, Leptidis S, Doevendans PA and De Windt LJ: HypoxamiRs: Regulators of cardiac hypoxia and energy metabolism. Trends Endocrinol Metab. 26:502–508. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Li B, Dasgupta C, Huang L, Meng X and Zhang L: MiRNA-210 induces microglial activation and regulates microglia-mediated neuroinflammation in neonatal hypoxic-ischemic encephalopathy. Cell Mol Immunoly. 17:976–991. 2020. View Article : Google Scholar

52 

Li R, Bao L, Hu W, Liang H and Dang X: Expression of miR-210 mediated by adeno-associated virus performed neuroprotective effects on a rat model of acute spinal cord injury. Tissue Cell. 57:22–33. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Camps C, Saini HK, Mole DR, Choudhry H, Reczko M, Guerra-Assunção JA, Tian YM, Buffa FM, Harris AL, Hatzigeorgiou AG, et al: Integrated analysis of microRNA and mRNA expression and association with HIF binding reveals the complexity of microRNA expression regulation under hypoxia. Mol Cancer. 13:282014. View Article : Google Scholar : PubMed/NCBI

55 

Zhang J, Ma J, Long K, Qiu W, Wang Y, Hu Z, Liu C, Luo Y, Jiang A, Jin L, et al: Overexpression of exosomal cardioprotective miRNAs mitigates hypoxia-induced H9c2 cells apoptosis. Int J Mol Sci. 18:7112017. View Article : Google Scholar : PubMed/NCBI

56 

Krown KA, Page MT, Nguyen C, Zechner D, Gutierrez V, Comstock KL, Glembotski CC, Quintana PJ and Sabbadini RA: Tumor necrosis factor alpha-induced apoptosis in cardiac myocytes. Involvement of the sphingolipid signaling cascade in cardiac cell death. J Clin Investig. 98:2854–2865. 1996. View Article : Google Scholar : PubMed/NCBI

57 

Chang L and Karin M: Mammalian MAP kinase signaling cascades. Nature. 410:37–40. 2001. View Article : Google Scholar : PubMed/NCBI

58 

Guo M, Shi JH, Wang PL and Shi DZ: Angiogenic growth factors for coronary artery disease: Current status and prospects. J Cardiovasc Pharmacol Ther. 23:130–141. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Kir D, Schnettler E, Modi S and Ramakrishnan S: Regulation of angiogenesis by microRNAs in cardiovascular diseases. Angiogenesis. 21:699–710. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Liberis A, Stanulov G, Ali EC, Hassan A, Pagalos A and Kontomanolis EN: Pre-eclampsia and the vascular endothelial growth factor: A new aspect. Clin Exp Obstet Gynecol. 43:9–13. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Ferrara N and Alitalo K: Clinical applications of angiogenic growth factors and their inhibitors. Nat Med. 5:1359–1364. 1999. View Article : Google Scholar : PubMed/NCBI

62 

Ylä-Herttuala S, Rissanen TT, Vajanto I and Hartikainen J: Vascular endothelial growth factors: Biology and current status of clinical applications in cardiovascular medicine. J Am Coll Cardiol. 49:1015–1026. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Fish JE and Srivastava D: MicroRNAs: Opening a new vein in angiogenesis research. Sci Signal. 2:pe12009. View Article : Google Scholar : PubMed/NCBI

64 

Zhang H, Hao J, Sun X, Zhang Y and Wei Q: Circulating pro-angiogenic micro-ribonucleic acid in patients with coronary heart disease. Interact Cardiovasc Thorac Surg. 27:336–342. 2018.PubMed/NCBI

65 

Xing Y, Hou J, Guo T, Zheng S, Zhou C, Huang H, Chen Y, Sun K, Zhong T, Wang J, et al: microRNA-378 promotes mesenchymal stem cell survival and vascularization under hypoxic-ischemic conditions in vitro. Stem Cell Res Ther. 5:1302014. View Article : Google Scholar : PubMed/NCBI

66 

Scheubel RJ, Holtz J, Friedrich I, Borgermann J, Kahrstedt S, Navarrete Santos A, Silber RE and Simm A: Paracrine effects of CD34 progenitor cells on angiogenic endothelial sprouting. Int J Cardiol. 139:134–141. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Lee DY, Deng Z, Wang CH and Yang BB: MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression. Proc Natl Acad Sci USA. 104:20350–20355. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Templin C, Volkmann J, Emmert MY, Mocharla P, Müller M, Kraenkel N, Ghadri JR, Meyer M, Styp-Rekowska B, Briand S, et al: Increased proangiogenic activity of mobilized CD34+ progenitor cells of patients with acute ST-segment-elevation myocardial infarction: Role of differential MicroRNA-378 expression. Arterioscler Thromb Vasc Biol. 37:341–349. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Naaijkens BA, van Dijk A, Meinster E, Kramer K, Kamp O, Krijnen PAJ, Niessen HWM and Juffermans LJM: Wistar rats from different suppliers have a different response in an acute myocardial infarction model. Res Vet Sci. 96:377–379. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Rajtik T, Carnicka S, Szobi A, Giricz Z, O-Uchi J, Hassova V, Svec P, Ferdinandy P, Ravingerova T and Adameova A: Data on necrotic and apoptotic cell death in acute myocardial ischemia/reperfusion injury: The effects of CaMKII and angiotensin AT1 receptor inhibition. Data Brief. 7:730–734. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Zhou R, Jia Y, Wang Y, Li Z, Qi J and Yang Y: Elevating miR-378 strengthens the isoflurane-mediated effects on myocardial ischemia-reperfusion injury in mice via suppression of MAPK1. Am J Transl Res. 13:2350–2364. 2021.PubMed/NCBI

72 

Zhang J, Chen F, Ma W and Zhang P: Suppression of long noncoding RNA NEAT1 attenuates hypoxia-induced cardiomyocytes injury by targeting miR-378a-3p. Gene. 731:1443242020. View Article : Google Scholar : PubMed/NCBI

73 

Dai R, Liu Y, Zhou Y, Xiong X, Zhou W, Li W, Zhou W and Chen M: Potential of circulating pro-angiogenic microRNA expressions as biomarkers for rapid angiographic stenotic progression and restenosis risks in coronary artery disease patients underwent percutaneous coronary intervention. J Clin Lab Anal. 1:e230132020.PubMed/NCBI

74 

Li H, Gao F, Wang X, Wu J, Lu K, Liu M, Li R, Ding L and Wang R: Circulating microRNA-378 levels serve as a novel biomarker for assessing the severity of coronary stenosis in patients with coronary artery disease. Biosci Rep. 39:BSR201820162019. View Article : Google Scholar : PubMed/NCBI

75 

Shen J, Chang C, Ma J and Feng Q: Potential of circulating proangiogenic MicroRNAs for predicting major adverse cardiac and cerebrovascular events in unprotected left main coronary artery disease patients who underwent coronary artery bypass grafting. Cardiology. 146:400–408. 2021. View Article : Google Scholar : PubMed/NCBI

76 

Lorenzen JM, Schauerte C, Hübner A, Kölling M, Martino F, Scherf K, Batkai S, Zimmer K, Foinquinos A, Kaucsar T, et al: Osteopontin is indispensible for AP1-mediated angiotensin II-related miR-21 transcription during cardiac fibrosis. Eur Heart J. 36:2184–2196. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Rosenberg M, Zugck C, Nelles M, Juenger C, Frank D, Remppis A, Giannitsis E, Katus HA and Frey N: Osteopontin, a new prognostic biomarker in patients with chronic heart failure. Circ Heart Fail. 1:43–49. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Kwee LC, Neely ML, Grass E, Gregory SG, Roe MT, Ohman EM, Fox KAA, White HD, Armstrong PW, Bowsman LM, et al: Associations of osteopontin and NT-proBNP with circulating miRNA levels in acute coronary syndrome. Physiol Genomics. 51:506–515. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Yuan J, Liu H, Gao W, Zhang L, Ye Y, Yuan L, Ding Z, Wu J, Kang L, Zhang X, et al: MicroRNA-378 suppresses myocardial fibrosis through a paracrine mechanism at the early stage of cardiac hypertrophy following mechanical stress. Theranostics. 8:2565–2582. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Xu T, Zhou Q, Che L, Das S, Wang L, Jiang J, Li G, Xu J, Yao J, Wang H, et al: Circulating miR-21, miR-378, and miR-940 increase in response to an acute exhaustive exercise in chronic heart failure patients. Oncotarget. 7:12414–12425. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Wang S, Long CL, Chen J, Cui WY, Zhang YF, Zhang H and Wang H: Pharmacological evidence: A new therapeutic approach to the treatment of chronic heart failure through SUR2B/Kir6.1 channel in endothelial cells. Acta Pharmacol Sin. 38:41–55. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Liu C, Lai Y, Pei J, Huang H, Zhan J, Ying S and Shen Y: Clinical and genetic analysis of KATP variants with heart failure risk in patients with decreased serum ApoA-I levels. J Clin Endocrinol Metab. 106:2264–2278. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Schiattarella GG and Hill J A: Inhibition of hypertrophy is a good therapeutic strategy in ventricular pressure overload. Circulation. 131:1435–1447. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Bernardo BC, Weeks KL, Pretorius L and McMullen JR: Molecular distinction between physiological and pathological cardiac hypertrophy: Experimental findings and therapeutic strategies. Pharmacol Ther. 128:191–227. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Ganesan J, Ramanujam D, Sassi Y, Ahles A, Jentzsch C, Werfel S, Leierseder S, Loyer X, Giacca M, Zentilin L, et al: MiR-378 controls cardiac hypertrophy by combined repression of mitogen-activated protein kinase pathway factors. Circulation. 127:2097–2106. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Chen YH, Zhong LF, Hong X, Zhu QL, Wang SJ, Han JB, Huang WJ and Ye BZ: Integrated analysis of circRNA-miRNA-mRNA ceRNA network in cardiac hypertrophy. Front Genet. 13:7816762022. View Article : Google Scholar : PubMed/NCBI

87 

Barut Z, Cabbar AT, Yilmaz SG, Akdeniz FT, Simsek MA, Capar B, Degertekin M, Dalan AB, Yerebakan H and Isbir T: Investigation of circulating miRNA-133, miRNA-26, and miRNA-378 as candidate biomarkers for left ventricular hypertrophy. In Vivo. 35:1605–1610. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Katz AM: Cardiomyopathy of overload. A major determinant of prognosis in congestive heart failure. N Engl J Med. 322:100–110. 1990. View Article : Google Scholar : PubMed/NCBI

89 

Borer JS, Truter S, Herrold EM, Falcone DJ, Pena M, Carter JN, Dumlao TF, Lee JA and Supino PG: Myocardial fibrosis in chronic aortic regurgitation: Molecular and cellular responses to volume overload. Circulation. 105:1837–1842. 2002. View Article : Google Scholar : PubMed/NCBI

90 

Creemers EE and Pinto YM: Molecular mechanisms that control interstitial fibrosis in the pressure-overloaded heart. Cardiovasc Res. 89:265–272. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Bishop JE and Lindahl G: Regulation of cardiovascular collagen synthesis by mechanical load. Cardiovasc Res. 42:27–44. 1999. View Article : Google Scholar : PubMed/NCBI

92 

Divakaran V and Mann DL: The emerging role of microRNAs in cardiac remodeling and heart failure. Circ Res. 103:1072–1083. 2008. View Article : Google Scholar : PubMed/NCBI

93 

Tatsuguchi M, Seok HY, Callis TE, Thomson JM, Chen JF, Newman M, Rojas M, Hammond SM and Wang DZ: Expression of microRNAs is dynamically regulated during cardiomyocyte hypertrophy. J Mol Cell Cardiol. 42:1137–1141. 2007. View Article : Google Scholar : PubMed/NCBI

94 

Moghiman T, Barghchi B, Esmaeili SA, Shabestari MM, Tabaee SS and Momtazi-Borojeni AA: Therapeutic angiogenesis with exosomal microRNAs: An effectual approach for the treatment of myocardial ischemia. Heart Fail Rev. 26:205–213. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Sun F, Zhuang Y, Zhu H, Wu H, Li D, Zhan L, Yang W, Yuan Y, Xie Y, Yang S, et al: LncRNA PCFL promotes cardiac fibrosis via miR-378/GRB2 pathway following myocardial infarction. J Mol Cell Cardiol. 133:188–198. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Zou Y, Li J, Ma H, Jiang H, Yuan J, Gong H, Liang Y, Guan A, Wu J, Li L, et al: Heat shock transcription factor 1 protects heart after pressure overload through promoting myocardial angiogenesis in male mice. J Mol Cell Cardiol. 51:821–829. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Lung B, Baron G, Butchart EG, Delahaye F, Gohlke-Bärwolf C, Levang OW, Tornos P, Vanoverschelde JL, Vermeer F, Boersma E, et al: A prospective survey of patients with valvular heart disease in Europe: The Euro heart survey on valvular heart disease. Eur Heart J. 24:1231–2143. 2003. View Article : Google Scholar : PubMed/NCBI

98 

Kupari M, Turto H and Lommi J: Left ventricular hypertrophy in aortic valve stenosis: Preventive or promotive of systolic dysfunction and heart failure? Eur Heart J. 26:1790–1796. 2005. View Article : Google Scholar : PubMed/NCBI

99 

Cramariuc D, Gerdts E, Davidsen ES, Segadal L and Matre K: Myocardial deformation in aortic valve stenosis: Relation to left ventricular geometry. Heart. 96:106–112. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Cioffi G, Faggiano P, Vizzardi E, Tarantini L, Cramariuc D, Gerdts E and de Simone G: Prognostic effect of inappropriately high left ventricular mass in asymptomatic severe aortic stenosis. Heart. 97:301–307. 2011. View Article : Google Scholar : PubMed/NCBI

101 

Mureddu GF, Cioffi G, Stefenelli C, Boccanelli A and de Simone G: Compensatory or inappropriate left ventricular mass in different models of left ventricular pressure overload: Comparison between patients with aortic stenosis and arterial hypertension. J Hypertens. 27:642–649. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Nagalingam RS, Sundaresan NR, Gupta MP, Geenen DL, Solaro RJ and Gupta M: A cardiac-enriched microRNA, miR-378, blocks cardiac hypertrophy by targeting Ras signaling. J Biol Chem. 288:11216–11232. 2013. View Article : Google Scholar : PubMed/NCBI

103 

Chen Z, Li C, Xu Y, Li Y, Yang H and Rao L: Circulating level of miR-378 predicts left ventricular hypertrophy in patients with aortic stenosis. PLoS One. 9:e1057022014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2023
Volume 28 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang H, Shi J, Wang J and Hu Y: MicroRNA‑378: An important player in cardiovascular diseases (Review). Mol Med Rep 28: 172, 2023
APA
Wang, H., Shi, J., Wang, J., & Hu, Y. (2023). MicroRNA‑378: An important player in cardiovascular diseases (Review). Molecular Medicine Reports, 28, 172. https://doi.org/10.3892/mmr.2023.13059
MLA
Wang, H., Shi, J., Wang, J., Hu, Y."MicroRNA‑378: An important player in cardiovascular diseases (Review)". Molecular Medicine Reports 28.3 (2023): 172.
Chicago
Wang, H., Shi, J., Wang, J., Hu, Y."MicroRNA‑378: An important player in cardiovascular diseases (Review)". Molecular Medicine Reports 28, no. 3 (2023): 172. https://doi.org/10.3892/mmr.2023.13059