Role of microRNAs in the pathogenesis of diabetic cardiomyopathy (Review)

  • Authors:
    • Xinyu Liu
    • Shixue Liu
  • View Affiliations

  • Published online on: January 12, 2017     https://doi.org/10.3892/br.2017.841
  • Pages: 140-145
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The morbidity of diabetes mellitus has been increasing annually. As a progressive metabolic disorder, chronic complications occur in the late stage of diabetes. In addition, cardiovascular diseases account for the major cause of morbidity and mortality among the diabetic population worldwide. Diabetic cardiomyopathy (DCM) is a type of diabetic heart disease. Patients with DCM show symptoms and signs of heart failure while no specific cause, such as coronary disease, hypertension, alcohol consumption, or other structural heart diseases has been identified. The pathogenesis of DCM is complex and has not been well understood until recently. MicroRNAs (miRs) belong to a novel family of highly conserved, short, non‑coding, single‑stranded RNA molecules that regulate transcriptional and post‑transcriptional gene expression. Furthermore, recent studies have demonstrated an association between miRs and DCM. In the current review, the role of miRs in the pathogenesis of DCM is summarized. It was concluded that miRs contribute to the regulation of cardiomyocyte hypertrophy, myocardial fibrosis, cardiomyocyte apoptosis, mitochondrial dysfunction, myocardial electrical remodeling, epigenetic modification and various other pathophysiological processes of DCM. These studies may provide novel insights into targets for prevention and treatment of the disease.

Introduction

According to data from the International Diabetes Federation, 382 million individuals presented with diabetes mellitus in 2013 and this number is expected to rise to 592 million by 2035 (1). As a progressive metabolic disorder, chronic complications occur in the late stage of diabetes, including atherosclerosis, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy and diabetic cardiomyopathy (DCM) (2). Among the vast array of long-term complications associated with diabetes, cardiovascular diseases account for the major cause of morbidity and mortality among the diabetic population worldwide (37).

The Framingham Heart Study demonstrated that diabetes was an independent risk factor of cardiovascular disease, including heart failure (8). The primary causes of diabetes-associated heart failure were believed to be coronary atherosclerosis and ischemia. However, in 1972, Rubler et al (9) reported autopsy data of four diabetic patients with heart failure, and no specific cause, such as coronary disease, hypertension, alcohol consumption, or other structural heart disease was identified. The authors subsequently introduced the term DCM. DCM is a newly identified disease and is considered to be completely different from coronary heart disease. Coronary heart disease usually results from atherosclerosis of the coronary artery and it is regarded as a type of macrovascular complication of diabetes. Diabetes promotes the onset and development of coronary heart disease together with numerous other factors, such as obesity, smoking and hyperlipidemia. However, the distinct features of DCM are cardiac hypertrophy and myocardial fibrosis in the absence of obvious pathogenic coronary abnormalities. It appears asymptomatic until the very late stage and left ventricular diastolic dysfunction with preserved systolic function is an early sign of DCM, while systolic dysfunction eventually occurs.

The pathogenesis of DCM is complex and has not been well understood until recently. Impaired calcium handling, altered metabolism, increased oxidative stress, remodeling of extracellular matrix (ECM), endothelial dysfunction and mitochondrial dysfunction have been found to participate in the pathogenesis of DCM (2,1014). A number of signaling proteins and pathways have been implicated in contributing to the development of DCM, including protein kinase C, nuclear factor-κB, peroxisome proliferator-activated receptor α, phosphatidylinositol 3-kinase (PI3K) and mitogen activated protein kinase (MAPK) signaling pathways (15,16).

In this context, microRNAs (miRs or miRNAs) are found to be important in the pathogenesis of DCM. miRs were initially described by Lee et al (17) in nematodes, Caenorhabditis elegans in 1993. As a novel family of highly conserved, short (~18–25 nucleotides), non-coding, single-stranded RNA molecules, miRs regulate transcriptional and post-transcriptional gene expression through binding to the 3′-untranslated region (3′-UTR) of their target mRNA (3). Given that miRs are crucially involved in numerous critical biological processes, including cell proliferation, apoptosis, necrosis, migration and differentiation, dysregulated miRs contribute to various human diseases, including diabetes and cardiovascular diseases (4,1820). miR-126, miR-17, miR-92a, miR-145, miR-155, miR-133 and miR-208a were identified to be associated with coronary artery disease; miR-1, miR-21, miR-208, miR-133a/b and miR-499 were identified as important in the pathogenesis of acute cardiac infarction. In addition, miRs associated with heart failure include miR-24, miR-125b, miR-195, miR-199a and miR-214 (2022).

Recent studies have demonstrated an association between miRs and DCM (2325). The expression level of miR in the hearts of patients with DCM was found to be different when compared with that of healthy individuals (25,26). Furthermore, analysis of miR expression levels in the hearts of various rat and mouse diabetic models also indicated the abnormal expression of miRNA. Further studies demonstrated that miRs contribute to numerous important pathophysiological processes of DCM, including cardiomyocyte hypertrophy, myocardial fibrosis, cardiomyocyte apoptosis, mitochondrial dysfunction, myocardial electrical remodeling and epigenetic modification (2732). The present review discusses the possible role of miRs in the pathogenesis of DCM regarding the above-mentioned processes.

miR involvement in the pathogenesis of DCM

miRs in cardiomyocyte hypertrophy

Cardiomyocyte hypertrophy is one of the distinct structural features of DCM. Studies have shown that various miRs were dysregulated and contributed to the pathogenesis of cardiomyocyte hypertrophy in DCM (29,3336). miR-30c, miR-133a, miR-150 and miR-373 were found to be downregulated in the heart of DCM, while miR-451 was found to be upregulated (29,3336).

miR-133 is abundantly expressed in heart tissue, and is known to regulate various physiological and pathophysiological events, including non-diabetic cardiac hypertrophy (3739). Hyperglycemia results in cardiac hypertrophy. A recent study reported that the expression level of miR-133a was reduced in cardiomyocytes treated with high levels of glucose, as well as in hypertrophic cardiac tissue samples from streptozotocin (STZ)-induced diabetic mice, and transfection of miR-133a mimics prevented altered gene expression and hypertrophic changes (33). Therefore, it was concluded that miR-133a participated in mediating glucose-induced cardiomyocyte hypertrophy in diabetes. Additionally, another study demonstrated that serum and glucocorticoid-regulated kinase 1 and insulin-like growth factor-1 (IGF-1) receptor may be involved in this process as potential targets of miR-133a (33).

Various anti- and pro-growth signaling pathways have been demonstrated to participate in cardiomyocyte hypertrophy, including the PI3K/AKT signaling pathway. p21-activated kinases (PAKs) and cell division control protein 42 homolog (Cdc42) are components of the PI3K/AKT signaling pathway, and PAKs are effectors of Cdc42. Myocardial Cdc42 and Pak1 mRNA and protein expression levels were found to be significantly increased in DCM rats with cardiac hypertrophy and in high glucose (HG)-treated cardiomyocytes, which was accompanied by a significant decrease in cardiac miR-30c expression levels in DCM rats (3.73-fold), patients with DCM (2.9-fold) and in HG-treated cardiomyocytes (3.5-fold) (35). Further investigation indicated that miR-30c possessed binding sites for the 3′-UTR and open reading frame (ORF) regions of Cdc42 and Pak1, and modulated Cdc42 and Pak1 expression levels in cardiomyocytes. In addition, miR-30c overexpression decreased HG-induced upregulation of Cdc42 and Pak1 and resulted in decreased expression levels of hypertrophic marker, atrial natriuretic peptide and a reduction in HG-treated cardiomyocyte cell size (35). These findings indicate that miR-30c exerts an anti-hypertrophic effect by inhibiting Cdc42 and Pak1 gene expression levels in DCM.

As a type of histone acetyl transferase, transcriptional co-activator, p300 has been confirmed to participate in the cardiomyocyte hypertrophy that is induced by pro-hypertrophic stimuli, particularly hyperglycemia (29). Further investigations found that the expression level of miR-150 was significantly reduced, whereas the expression level of p300 was markedly elevated, concomitant with cardiomyocyte hypertrophy, in the hypertrophic hearts of diabetic rats and in neonatal rat cardiomyocytes exposed to high levels of glucose (29). In addition, a luciferase reporter activity assay indicated that miR-150 functioned directly with the 3′-UTR of p300 and miR-150 mimics prevented glucose-induced cardiomyocyte hypertrophy (29). Thus, it was concluded that miR-150 was important in p300-mediated cardiomyocyte hypertrophy in response to hyperglycemia.

miR-373 has also been demonstrated to be involved in the pathogenesis of hyperglycemia-induced cardiac hypertrophy. It was found to be downregulated in heart samples of STZ-induced diabetic mice, and exposure of neonatal rat cardiomyocytes to glucose and transfection with miR-373 mimic demonstrated increased expression levels of miR-373 and cell size, indicating a strong involvement of miR-373 in glucose-induced cardiomyocyte hypertrophy (36). In addition, the study revealed that miR-373 was transcriptionally regulated by p38 MAPK and that its anti-hypertrophic effects may be mediated by targeting the hypertrophic protein, myocyte enhancer factor 2C (36).

Triglyceride accumulation and excess supply of saturated fatty acids, such as palmitic acid, have been implicated in the induction of cardiac hypertrophy in diabetes. miR-451 expression levels were observed to be significantly elevated in diet-induced obesity (DIO) mouse hearts with hypertrophy and in neonatal rat cardiomyocytes stimulated with palmitate (34). In addition, high-fat diet-induced cardiac hypertrophy and contractile reserves were ameliorated in cardiomyocyte-specific miR-451 knockout mice compared with the control (34). As an important component of the liver kinase B1 (LKB1)/adenosine monophosphate activated protein kinase (AMPK) signaling pathway, calcium-binding protein 39 (Cab39) was identified to be a direct miR-451 target in neonatal rat cardiac myocytes. Further experiments indicated that protein expression levels of Cab39 and phosphorylated AMPK were increased and phosphorylated mammalian target of rapamycin (mTOR) was reduced in cardiomyocyte-specific miR-451 knockout mouse hearts compared with control mouse hearts, demonstrating that miR-451 was involved in DCM via suppression of the LKB1/AMPK signaling pathway (34).

miRs in myocardial fibrosis

Myocardial fibrosis is another main cause of DCM. Abnormally elevated ECM deposition, in particular collagen deposition, increases myocardial stiffness, leads to irreversible tissue damage and finally results in myocardial fibrosis (16).

In addition to cardiac hypertrophy, miR-133a was identified to be involved in the pathogenesis of diabetes-induced myocardial fibrosis. The expression level of miR-133a was decreased in the hearts of STZ-induced diabetic mice, accompanied by an increase in the transcriptional co-activator, p300 and in major markers of fibrosis (transforming growth factor-β1, connective tissue growth factor, fibronectin and collagen 1 α1V), as well as increased focal cardiac fibrosis, as measured by Masson's trichome stain (28). Furthermore, miR-133a overexpression prominently alleviates cardiac fibrosis as observed by assessment of major fibrosis markers and microscopic examination, indicating miR-133a as a potential therapeutic target for combatting cardiac fibrosis (28).

The peripheral blood level of miR-21 has been demonstrated as a biomarker for myocardial fibrosis (40). A recent study showed that miR-21 was upregulated in rat cardiac fibroblasts in response to high levels of glucose, accompanied by promoted fibroblast proliferation and collagen synthesis (41). In addition, dual specific phosphatase 8 (DUSP8) was identified to be a direct target of miR-21; the expression of DUSP8 was suppressed by miR-21, which promoted HG-induced cardiac fibrosis by affecting the activity of the c-Jun N-terminal kinase/stress activated protein kinase and p38 signaling pathways (41).

Thus, miR-133a and miR-21 are dysregulated by HG stimulation, leading to myocardial fibrosis in DCM. Interventions focusing on the expression levels of these miRs may result in novel concepts for improving DCM remodeling.

miRs in cardiomyocyte apoptosis and mitochondrial dysfunction

Various miRs have been demonstrated to be involved in DCM-associated cardiomyocyte apoptosis and mitochondrial dysfunction, including miR-34a, miR-1, miR-206, mi-195 and mi-30d.

High levels of glucose may induce cardiomyocyte apoptosis and thus contribute to the pathogenesis of DCM; miR-34a was found to be involved in this process. Upregulation of miR-34a expression levels and a decrease in the B cell leukemia/lymphoma 2 (Bcl-2) expression level were observed in the rat H9C2 cardiomyocyte cell line when exposed to HG, while apoptosis of H9C2 cells was significantly increased (42). Furthermore, treatment with miR-34a mimics significantly decreased the Bcl-2 expression level and promoted HG-induced apoptotic changes in H9C2 cells, whereas treatment with an miR-34a inhibitor markedly increased the Bcl-2 expression level and prevented H9C2 cell apoptosis, indicating that miR-34a was critical in the HG-induced decrease of the pro-apoptosis protein, Bcl-2 expression level and subsequent cardiomyocyte apoptosis (42).

The molecular chaperone heat shock protein 60 (Hsp60) is an important anti-apoptotic protein, which may regulate the Bcl-2 family. Reduced expression levels of the Hsp60 protein were observed in the diabetic rat myocardium and HG-treated neonatal rat ventricular cardiomyocytes, which was accompanied by significant upregulation of miR-1 and miR-206 (43). Further studies then demonstrated that rat miR-1 and miR-206 negatively regulated Hsp60 expression by directly targeting the 3′-UTR of Hsp60 mRNA, and miR-1 and miR-206 mediated their effects on H9C2 cell apoptosis via Hsp60 (43). These findings indicated that miR-1 and miR-206 modulate the expression of their common target, Hsp60 and consequently mediated HG-induced apoptosis of cardiomyocytes.

In addition, miR-1 was found to mediate apoptosis of HG-treated H9C2 cells through regulating another potential target, IGF-1, which is proposed to be an anti-apoptosis factor (44). It was observed that H9C2 cells exposed to HG levels exhibited increased miR-1 expression levels, decreased mitochondrial membrane potential, increased cytochrome c release and increased apoptosis; however, these consequences were detected to be blocked by IGF-1 (44).

Another study demonstrated that the level of miR-195 expression was increased and expression levels of its target proteins (Bcl-2 and sirtuin 1) were decreased in STZ-induced type 1 and db/db type 2 diabetic mouse hearts (45). Upregulation of miR-195 in diabetic hearts was associated with oxidative stress, apoptosis, myocardial hypertrophy and dysfunction, as well as a reduction in coronary blood flow while silencing of miR-195 reduces oxidative damage, apoptosis and hypertrophy, and restores coronary blood flow in diabetic hearts, with a concurrent upregulation of Bcl-2 and sirtuin 1, leading to an improvement in myocardial function (45). This study validated the role of miR-195 in promoting apoptosis in the DCM heart, as well as in other pathophysiological changes (45).

Pyroptosis is pro-inflammatory programmed cell death and it is another type of cell death that is different from apoptosis or necrosis (46). HG may induce cardiomyocyte pyroptosis and miR-30d was observed to be involved in this process. It was revealed that miR-30d expression was substantially increased in STZ-induced diabetic rats, as well as in HG-treated cardiomyocytes (30). Furthermore, upregulation of miR-30d promoted cardiomyocyte pyroptosis in DCM by directly targeting forkhead box O3, which resulted in suppression of the expression of its downstream protein, apoptosis repressor with caspase recruitment domain and upregulated expression of inflammatory molecules, including caspase-1, interleukin (IL)-1β and IL-18, and finally led to pyroptosis of cardiomyocyte (30).

Dysfunction of mitochondria also contributes to the pathogenesis of DCM and miR-141 was found to participate in this process. The expression level of miR-141 was significantly upregulated in the hearts of STZ-induced diabetic mice. Furthermore, through regulating its potential target, solute carrier family 25 member 3, which provides inorganic phosphate to the mitochondrial matrix and is essential for ATP production as an inner mitochondrial membrane phosphate transporter, overexpression of miR-141 was indicated to decrease inorganic phosphate transport and exert functional implications for mitochondrial ATP production (47).

miRs and other pathophysiological processes of DCM

miRs are also involved in the pathogenesis of DCM through participating in various pathophysiological processes, such as myocardial electrical remodeling and epigenetic modification (27,31).

A significant increase in the level of miR-301 expression and reduction of the voltage gated potassium channel, Kv4.2 expression level were observed in the diabetic (db/db mice) ventricles and miR-301 was validated to modulate Kv4.2 by direct binding on its 3′-UTR (31). Kv4.2 is important in maintaining the cardiac repolarization reserve, and the depletion of repolarization reserve was further identified in the diabetic hearts, elucidating that miR-301 mediated DCM by regulating myocardial electrical remodeling (31).

DNA methylation is an important aspect of epigenetic modification. In addition to its role in mediating myocardial hypertrophy and fibrosis, miR-133a was found to contribute to hyperglycemia-mediated DNA hypermethylation by regulating the expression levels of DNA methyl transferases, which catalyze DNA methylation. It was observed that the expression of miR-133a was attenuated while DNA methyl transferase (Dnmt)-1 and −3b were induced in Ins2+/− Akita hearts, and overexpression of miR-133a inhibits, but silencing of miR-133a induces, Dnmt-1, −3a and −3b, demonstrating the involvement of miR-133a in the regulation of DNA methylation (27).

Conclusion

In conclusion, miRs are crucial in the pathogenesis of DCM by regulating cardiomyocyte hypertrophy, myocardial fibrosis, cardiomyocyte apoptosis, mitochondrial dysfunction, myocardial electrical remodeling, epigenetic modification and various other pathophysiological processes, as shown in Table I and Fig. 1. Furthermore, numerous studies have demonstrated that interventions with the expression levels of associated miRNs may improve the pathophysiological process of DCM, providing novel insights into targets for the prevention and treatment of DCM (28,29,34,35,45). However, those studies were limited to the expression changes of miRs in heart tissue samples. To the best of our knowledge, circulating miRNs have not yet been identified to be specifically dysregulated in DCM. Further studies and clinical observations are required to identify circulating miRs as biomarkers for early prediction and diagnosis of DCM.

Table I.

miRs that are dysregulated in DCM and their roles.

Table I.

miRs that are dysregulated in DCM and their roles.

Author, yearmiRsExpressionType of model investigatedPotential targetsRole in DCMRefs.
Chavali et al, 2012;miR-133aDownregulatedCardiac tissues of STZ-induced and Ins2+/−SGK1 and IGF1R;Antihypertrophic effect; regulating(27,28,33)
Chen et al, 2014; and Feng et al, 2010 Akita diabetic mice; neonatal rat myocytes exposed to high levels of glucoseTGFβ1, FGF1, CTGFDNA methylation; protection against myocardial fibrosis
Duan et al, 2013miR-150DownregulatedHearts of STZ diabetic SD rats; HG-treated primary neonatal rat cardiomyocytesp300Antihypertrophic effect(29)
Li et al, 2014miR-30dUpregulatedHearts of STZ-induced diabetic rats and high glucose-treated cardiomyocytesFoxo3aPromoting pyroptosis(30)
Panguluri et al, 2013miR-301UpregulatedRight ventricle and left ventricle of db/db miceKv4.2Inducing depletion of repolarization reserve(31)
Kuwabara et al, 2015miR-451UpregulatedDIO mouse hearts; neonatal rat cardiomyocytes stimulated with palmitateCab39Prohypertrophic effect(34)
Raut et al, 2015miR-30cDownregulatedHearts of Wistar rats fed with HFD and low-dose STZ; cardiac tissue samples from patients with DCM; HG-treated rat cardiomyocyte cell line (H9C2)Cdc42 and Pak1Antihypertrophic effect(35)
Shen et al, 2011miR-373DownregulatedHeart samples of STZ-induced diabetic miceMEF2CAntihypertrophic effect(36)
Liu et al, 2014miR-21UpregulatedRat cardiac fibroblasts treated with a high level of glucoseDUSP8Promoting myocardial fibrosis(41)
Zhao et al, 2013miR-34aUpregulatedHigh glucose-treated rat cardiomyocyte H9C2 cell lineBcl-2Promoting apoptosis(42)
Shan et al, 2010miR-1 and miR-206UpregulatedMyocardium of STZ-induced diabetic rat and high glucose-treated neonatal rat ventricular cardiomyocytesHsp60Promoting apoptosis(43)
Yu et al, 2008miR-1UpregulatedH9C2 cells exposed to high glucoseIGF-1Inducing mitochondrial dysfunction, cytochrome c release and apoptosis(44)
Zheng et al, 2015miR-195UpregulatedHearts of STZ-induced type 1 and db/db type 2 diabetic mice; high glucose-treated rat cardiomyocytesBcl-2 and sirtuin 1Promoting oxidative stress, apoptosis, myocardial hypertrophy and dysfunction(45)
Baseler et al, 2012miR-141UpregulatedHearts of STZ-induced diabetic Friend Virus B miceSlc25a3Decreasing mitochondrial ATP production(47)

[i] STZ, streptozotocin; SGK1, serum and glucocorticoid-regulated kinase 1; IGF1R, insulin-like growth factor-1 receptor; TGFβ1, transforming growth factor-β1; FGF1, fibroblast growth factor 1; CTGF, connective tissue growth factor; HFD, high fat diet; DCM, diabetic cardiomyopathy; Cdc42, cell division control protein 42 homolog; Pak1, p21-activated kinase 1; HG, high glucose; p300, transcriptional co-activator p300; DIO, diet-induced obesity; Cab39, calcium-binding protein 39; MEF2C, myocyte enhancer factor 2C; DUSP8, dual specific phosphatase 8; Bcl-2, B cell leukemia/lymphoma 2; Hsp60, heat shock protein 60; IGF-1, insulin-like growth factor 1; Foxo3a, forkhead box O3; Slc25a3, solute carrier family 25 member 3; Kv4.2, voltage gate potassium channel 4.2.

References

1 

Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U and Shaw JE: Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 103:137–149. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Wegner M, Neddermann D, Piorunska-Stolzmann M and Jagodzinski PP: Role of epigenetic mechanisms in the development of chronic complications of diabetes. Diabetes Res Clin Pract. 105:164–175. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Rawal S, Manning P and Katare R: Cardiovascular microRNAs: As modulators and diagnostic biomarkers of diabetic heart disease. Cardiovasc Diabetol. 13:442014. View Article : Google Scholar : PubMed/NCBI

4 

Zhou Q, Lv D, Chen P, Xu T, Fu S, Li J and Bei Y: MicroRNAs in diabetic cardiomyopathy and clinical perspectives. Front Genet. 5:1852014. View Article : Google Scholar : PubMed/NCBI

5 

Beckman JA, Creager MA and Libby P: Diabetes and atherosclerosis: Epidemiology, pathophysiology, and management. JAMA. 287:2570–2581. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Chavali V, Tyagi SC and Mishra PK: Predictors and prevention of diabetic cardiomyopathy. Diabetes Metab Syndr Obes. 6:151–160. 2013.PubMed/NCBI

7 

Hayat SA, Patel B, Khattar RS and Malik RA: Diabetic cardiomyopathy: Mechanisms, diagnosis and treatment. Clin Sci (Lond). 107:539–557. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Kannel WB and McGee DL: Diabetes and glucose tolerance as risk factors for cardiovascular disease: The Framingham study. Diabetes Care. 2:120–126. 1979. View Article : Google Scholar : PubMed/NCBI

9 

Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW and Grishman A: New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol. 30:595–602. 1972. View Article : Google Scholar : PubMed/NCBI

10 

Trachanas K, Sideris S, Aggeli C, Poulidakis E, Gatzoulis K, Tousoulis D and Kallikazaros I: Diabetic cardiomyopathy: From pathophysiology to treatment. Hellenic J Cardiol. 55:411–421. 2014.PubMed/NCBI

11 

Yilmaz S, Canpolat U, Aydogdu S and Abboud HE: Diabetic Cardiomyopathy; Summary of 41 Years. Korean Circ J. 45:266–272. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Falcão-Pires I and Leite-Moreira AF: Diabetic cardiomyopathy: Understanding the molecular and cellular basis to progress in diagnosis and treatment. Heart Fail Rev. 17:325–344. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Singh GB, Sharma R and Khullar M: Epigenetics and diabetic cardiomyopathy. Diabetes Res Clin Pract. 94:14–21. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Asrih M and Steffens S: Emerging role of epigenetics and miRNA in diabetic cardiomyopathy. Cardiovasc Pathol. 22:117–125. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Liu JW, Liu D, Cui KZ, Xu Y, Li YB, Sun YM and Su Y: Recent advances in understanding the biochemical and molecular mechanism of diabetic cardiomyopathy. Biochem Biophys Res Commun. 427:441–443. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Huynh K, Bernardo BC, McMullen JR and Ritchie RH: Diabetic cardiomyopathy: Mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther. 142:375–415. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

18 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Bauersachs J and Thum T: Biogenesis and regulation of cardiovascular microRNAs. Circ Res. 109:334–347. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Udali S, Guarini P, Moruzzi S, Choi SW and Friso S: Cardiovascular epigenetics: From DNA methylation to microRNAs. Mol Aspects Med. 34:883–901. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Fichtlscherer S, Zeiher AM, Dimmeler S and Sessa WC: Circulating microRNAs: Biomarkers or mediators of cardiovascular diseases? Arterioscler Thromb Vasc Biol. 31:2383–2390. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, Weber M, Hamm CW, Röxe T, Müller-Ardogan M, et al: Circulating microRNAs in patients with coronary artery disease. Circ Res. 107:677–684. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Figueira MF, Monnerat-Cahli G, Medei E, Carvalho AB, Morales MM, Lamas ME, da Fonseca RN and Souza-Menezes J: MicroRNAs: Potential therapeutic targets in diabetic complications of the cardiovascular and renal systems. Acta Physiol (Oxf). 211:491–500. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Diao X, Shen E, Wang X and Hu B: Differentially expressed microRNAs and their target genes in the hearts of streptozotocin-induced diabetic mice. Mol Med Rep. 4:633–640. 2011.PubMed/NCBI

25 

Rawal S, Ram TP, Coffey S, Williams MJ, Saxena P, Bunton RW, Galvin IF and Katare R: Differential expression pattern of cardiovascular microRNAs in the human type-2 diabetic heart with normal ejection fraction. Int J Cardiol. 202:40–43. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Nandi SS, Duryee MJ, Shahshahan HR, Thiele GM, Anderson DR and Mishra PK: Induction of autophagy markers is associated with attenuation of miR-133a in diabetic heart failure patients undergoing mechanical unloading. Am J Transl Res. 7:683–696. 2015.PubMed/NCBI

27 

Chavali V, Tyagi SC and Mishra PK: MicroRNA-133a regulates DNA methylation in diabetic cardiomyocytes. Biochem Biophys Res Commun. 425:668–672. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Chen S, Puthanveetil P, Feng B, Matkovich SJ, Dorn GW II and Chakrabarti S: Cardiac miR-133a overexpression prevents early cardiac fibrosis in diabetes. J Cell Mol Med. 18:415–421. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Duan Y, Zhou B, Su H, Liu Y and Du C: miR-150 regulates high glucose-induced cardiomyocyte hypertrophy by targeting the transcriptional co-activator p300. Exp Cell Res. 319:173–184. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Li X, Du N, Zhang Q, Li J, Chen X, Liu X, Hu Y, Qin W, Shen N, Xu C, et al: MicroRNA-30d regulates cardiomyocyte pyroptosis by directly targeting foxo3a in diabetic cardiomyopathy. Cell Death Dis. 5:e14792014. View Article : Google Scholar : PubMed/NCBI

31 

Panguluri SK, Tur J, Chapalamadugu KC, Katnik C, Cuevas J and Tipparaju SM: MicroRNA-301a mediated regulation of Kv4.2 in diabetes: Identification of key modulators. PLoS One. 8:e605452013. View Article : Google Scholar : PubMed/NCBI

32 

Yu M, Liu Y, Zhang B, Shi Y, Cui L and Zhao X: Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of streptozotocin-induced diabetic mice. Cardiovasc Pathol. 24:375–381. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Feng B, Chen S, George B, Feng Q and Chakrabarti S: miR133a regulates cardiomyocyte hypertrophy in diabetes. Diabetes Metab Res Rev. 26:40–49. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kuwabara Y, Horie T, Baba O, Watanabe S, Nishiga M, Usami S, Izuhara M, Nakao T, Nishino T, Otsu K, et al: MicroRNA-451 exacerbates lipotoxicity in cardiac myocytes and high-fat diet-induced cardiac hypertrophy in mice through suppression of the LKB1/AMPK pathway. Circ Res. 116:279–288. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Raut SK, Kumar A, Singh GB, Nahar U, Sharma V, Mittal A, Sharma R and Khullar M: miR-30c Mediates Upregulation of Cdc42 and Pak1 in Diabetic Cardiomyopathy. Cardiovasc Ther. 33:89–97. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Shen E, Diao X, Wang X, Chen R and Hu B: MicroRNAs involved in the mitogen-activated protein kinase cascades pathway during glucose-induced cardiomyocyte hypertrophy. Am J Pathol. 179:639–650. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Liu N, Bezprozvannaya S, Williams AH, Qi X, Richardson JA, Bassel-Duby R and Olson EN: MicroRNA-133a regulates cardiomyocyte proliferation and suppresses smooth muscle gene expression in the heart. Genes Dev. 22:3242–3254. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Barringhaus KG and Zamore PD: MicroRNAs: Regulating a change of heart. Circulation. 119:2217–2224. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Carè A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, Gu Y, Dalton ND, et al: MicroRNA-133 controls cardiac hypertrophy. Nat Med. 13:613–618. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, Dean DB and Zhang C: MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ Res. 100:1579–1588. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Liu S, Li W, Xu M, Huang H, Wang J and Chen X: Micro-RNA 21Targets dual specific phosphatase 8 to promote collagen synthesis in high glucose-treated primary cardiac fibroblasts. Can J Cardiol. 30:1689–1699. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Zhao F, Li B, Wei YZ, Zhou B, Wang H, Chen M, Gan XD, Wang ZH and Xiong SX: MicroRNA-34a regulates high glucose-induced apoptosis in H9c2 cardiomyocytes. J Huazhong Univ Sci Technolog Med Sci. 33:834–839. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Shan ZX, Lin QX, Deng CY, Zhu JN, Mai LP, Liu JL, Fu YH, Liu XY, Li YX, Zhang YY, et al: miR-1/miR-206 regulate Hsp60 expression contributing to glucose-mediated apoptosis in cardiomyocytes. FEBS Lett. 584:3592–3600. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Yu XY, Song YH, Geng YJ, Lin QX, Shan ZX, Lin SG and Li Y: Glucose induces apoptosis of cardiomyocytes via microRNA-1 and IGF-1. Biochem Biophys Res Commun. 376:548–552. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Zheng D, Ma J, Yu Y, Li M, Ni R, Wang G, Chen R, Li J, Fan GC, Lacefield JC, et al: Silencing of miR-195 reduces diabetic cardiomyopathy in C57BL/6 mice. Diabetologia. 58:1949–1958. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, et al: Nomenclature Committee on Cell Death 2009: Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 16:3–11. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Baseler WA, Thapa D, Jagannathan R, Dabkowski ER, Croston TL and Hollander JM: miR-141 as a regulator of the mitochondrial phosphate carrier (Slc25a3) in the type 1 diabetic heart. Am J Physiol Cell Physiol. 303:C1244–C1251. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 6 Issue 2

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu X and Liu X: Role of microRNAs in the pathogenesis of diabetic cardiomyopathy (Review). Biomed Rep 6: 140-145, 2017
APA
Liu, X., & Liu, X. (2017). Role of microRNAs in the pathogenesis of diabetic cardiomyopathy (Review). Biomedical Reports, 6, 140-145. https://doi.org/10.3892/br.2017.841
MLA
Liu, X., Liu, S."Role of microRNAs in the pathogenesis of diabetic cardiomyopathy (Review)". Biomedical Reports 6.2 (2017): 140-145.
Chicago
Liu, X., Liu, S."Role of microRNAs in the pathogenesis of diabetic cardiomyopathy (Review)". Biomedical Reports 6, no. 2 (2017): 140-145. https://doi.org/10.3892/br.2017.841