miR-106a promotes cardiac hypertrophy by targeting mitofusin 2

https://doi.org/10.1016/j.yjmcc.2016.08.016Get rights and content

Highlights

  • As a new molecular target miR-106a induces hypertrophy in cardiomyocytes.

  • Mfn2 as a cardiac protective factor alleviates hypertrophy.

  • miR-106a promotes cardiac hypertrophy by inhibiting Mfn2.

Abstract

Pathological cardiac hypertrophy is a main factor leading to heart failure and associated sudden death. Improved understanding of the underlying molecular mechanisms should aid better treatment of the disease. This study aimed to test our hypothesis that a microRNA miR-106a played an important role in the development of cardiac hypertrophy through targeting mitofusin 2 (Mfn2), a mitochondrial fusion protein known to be critical in regulating cardiac function. miR-106a was robustly upregulated in hypertrophied myocardium both in vivo and in vitro. Forced transient expression of miR-106a in otherwise healthy cardiomyocytes induced the hypertrophic phenotypes resembling those produced by angiotensin II (AngII) exposure. Knockdown of miR-106a by its specific inhibitor nearly completely reversed the hypertrophic phenotypes induced by AngII pretreatment and pressure overload. On the other hand, Mfn2 was markedly downregulated in hypertrophic heart and cardiomyocytes, which was in reciprocal to expression of miR-106a. Mfn2 was experimentally validated as a direct target gene for miR-106a. Overexpression of Mfn2 counteracted the hypertrophic responses induced by miR-106a, whereas silence of Mfn2 by its siRNA abolished the anti-hypertrophic property of miR-106a inhibitor. Furthermore, overexpression of Mfn2 alleviated the hypertrophic phenotypes induced by AngII in cultured cardiomyocytes, while Mfn2 siRNA alone was able to induce hypertrophic changes in cultured cardiomyocytes. Moreover, AngII and miR-106a treatment cultured cardiomyocytes mitochondria presented cristae defects, considerable depolarization of mitochondrial membrane and increased ROS production. These alterations were reversed by miR-106a inhibitor or overexpression of Mfn2. Taken together, our findings indicate miR-106a as an important factor to promote hypertrophic progress and suggest miR-106a as a new molecular target for the treatment of pathological hypertrophy. The present study also uncovered a novel relationship between miR-106a and Mfn2, with Mfn2 as a downstream signaling mediator of miR-106a.

Introduction

Cardiac hypertrophy was thought to be an adaptive response to various stresses to maintain normal cardiac functions. It is now believed that cardiac hypertrophy in many cases is a pathological process, as it can lead to heart failure, arrhythmia and even sudden death [1], [2]. Cardiac hypertrophy is characterized by cardiomyocyte growth in size at the cellular level, increased protein synthesis and re-expression of many fetal genes at the molecular level, and increased left ventricular (LV) wall thickness and impaired cardiac function at the organ level [3]. It is known that cardiac hypertrophy can be induced by a variety of cardiovascular disorders such as long-standing hypertension, valvular insufficiency and stenosis, myocarditis, and diabetic cardiomyopathy.

An increasing body of evidence indicates that microRNAs (miRNAs) serve as an important layer of the regulatory network in cardiovascular disease including cardiac hypertrophy. miRNAs are a class of small, ~ 22-nucleotides-long, non-coding RNAs that are believed to regulate the expression of up to 30% of human genes by binding to the 3′-UTR of mRNAs [4], [5]. By regulating expression of protein-coding genes, miRNAs can often result in well-defined phenotypes of pathophysiological processes in humans and animal models. For example, ectopic expression of miR-1, miR-328 and miR-21 has been reported to be critically involved in acute myocardial infarction, arrhythmogenesis and cardiac hypertrophy [6], [7], [8]. The miR-17-92 cluster has been shown to participate in cardiac ischemic/reperfusion injury [9], and deregulation of miR-17-92 causes lethal hypertrophic cardiomyopathy and arrhythmogenesis [10]. Moreover, these miRNAs have also been found to be required for cardiomyocyte proliferation in postnatal and adult hearts [11]. In our pilot study aiming to investigate the role of the miR-17-5p seed family (the miRNA sharing the same seed site as miR-17-5p) in cardiac hypertrophy, we found that miR-106a was substantially up-regulated. Our theoretical analysis further indicated that this miR-106a has the potential to target a gene called Mfn2 which is reportedly important in regulating cardiac hypertrophy [12], [13]. However, the role of miR-106a in the heart has not been studied.

Mfn2 is a key member of mitofusin protein family, which is located in outer membrane of mitochondrial and acts to maintain the structure of mitochondrial [14]. Mfn2 is highly expressed in tissues and cells with high metabolism rates such as heart, skeletal muscle, brain, kidney, and liver [15], [16], [17], [18], [19]. It promotes mitochondrial fusion to interchange mitochondrial DNA or energy with nearby mitochondria. It also interlinks mitochondrial and sarcoplasmic reticulum to promote mitochondrial respiration [20], [21]. Mfn2 is known as a cell proliferation suppressor gene, probably by inhibiting the MAPK/ERK pathway [15], [22]. Recent studies indicate that Mfn2 deficiency is involved in cardiovascular disease [17], [23], [24].

On the basis of these facts, we hypothesized that miR-106a might be involved in hypertrophic growth of the heart through targeting Mfn2. This study was designed to examine this notion in a mouse model of cardiac hypertrophy and a cellular model of cardiomyocyte hypertrophy.

Section snippets

Animals

C57BL/6 mice were used. The animals were kept under standard animal room conditions (temperature 21 ± 1 °C; humidity 55–60%) with food and water continuously available for 1 week before the experiments. All experimental procedures were conformed to the NIH guidelines and were approved by the Institutional Animal Care and Use Committee of Harbin Medical University.

Pressure-overload cardiac hypertrophy

The pressure-overload model of cardiac hypertrophy was created by transverse aortic constriction (TAC). Adult mice (C57BL/6), weighing

Up-regulation of miR-106a correlates with hypertrophic phenotypes in vivo and in vitro

C57BL/6 mice underwent transverse aortic constriction operation to induce cardiac hypertrophy or sham operation for negative control. The mice were kept in the same conditions with food and water continuously available for three weeks. Echocardiography analysis was implemented to measure left vLV wall thickness. Echocardiography analysis showed that LV wall was thicker in the hypertrophied hearts after three weeks of TAC mice compared with the hearts of sham mice (Fig. 1A). The hearts were

Discussion

To date, no < 1500 human miRNAs have been identified. Though they are theoretically predicted to target thousands of protein-coding genes involving a wide spectrum of biological and pathophysiological processes, experimental characterization of their functions has been limited to a small portion of these known miRNAs. In the past years, a number of miRNAs have been reported to participate in cardiac hypertrophy. These findings have greatly advanced our understanding of the molecular mechanisms

Conclusion

Collectively, we presented the first evidence that miR-106a overexpression is sufficient to induce hypertrophic growth via directly targeting Mfn2. In light of the fact that knockdown of miR-106a was able to reverse the hypertrophic alterations, together with the observation that miR-106a was abnormally upregulated, it may be speculated that miR-106a is a new molecular target for the treatment of pathological hypertrophy and other possible cardiac maladies associated with miR-106a

Conflict of interest

None declared.

Sources of funding

This work was supported by National Natural Science fund of China (81473213).

Acknowledgements

We gratefully acknowledge the assistance of Prof. Zhiguo Wang for the expert proofreading of our manuscript.

References (52)

  • S.B. Ong et al.

    Mitochondrial fusion and fission proteins as novel therapeutic targets for treating cardiovascular disease

    Eur. J. Pharmacol.

    (2015)
  • J.A. Hill et al.

    Cardiac plasticity

    N. Engl. J. Med.

    (2008)
  • B.C. Berk et al.

    ECM remodeling in hypertensive heart disease

    J. Clin. Invest.

    (2007)
  • J. Heineke et al.

    Regulation of cardiac hypertrophy by intracellular signalling pathways

    Nat. Rev. Mol. Cell Biol.

    (2006)
  • S. Gu et al.

    Biological basis for restriction of microRNA targets to the 3′ untranslated region in mammalian mRNAs

    Nat. Struct. Mol. Biol.

    (2009)
  • S. Roy et al.

    MicroRNA expression in response to murine myocardial infarction: mir-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue

    Cardiovasc. Res.

    (2009)
  • B. Yang et al.

    The muscle-specific microRNA mir-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2

    Nat. Med.

    (2007)
  • L.S. Danielson et al.

    Cardiovascular dysregulation of mir-17-92 causes a lethal hypertrophic cardiomyopathy and arrhythmogenesis

    FASEB J.

    (2013)
  • J. Chen et al.

    mir-17-92 cluster is required for and sufficient to induce cardiomyocyte proliferation in postnatal and adult hearts

    Circ. Res.

    (2013)
  • H. Yu et al.

    Mitofusin 2 inhibits angiotensin II-induced myocardial hypertrophy

    J. Cardiovasc. Pharmacol. Ther.

    (2011)
  • K.H. Chen et al.

    Dysregulation of HSG triggers vascular proliferative disorders

    Nat. Cell Biol.

    (2004)
  • C. Chen et al.

    Contribution of neural cell death to depressive phenotypes of streptozotocin-induced diabetic mice

    Dis. Model Mech.

    (2014)
  • M. Song et al.

    Super-suppression of mitochondrial reactive oxygen species signaling impairs compensatory autophagy in primary mitophagic cardiomyopathy

    Circ. Res.

    (2014)
  • F.B. Hickey et al.

    IHG-1 increases mitochondrial fusion and bioenergetic function

    Diabetes

    (2014)
  • A. Sood et al.

    A mitofusin-2-dependent inactivating cleavage of OPA1 links changes in mitochondria cristae and ER contacts in the postprandial liver

    Proc. Natl. Acad. Sci. U. S. A.

    (2014)
  • K.N. Papanicolaou et al.

    Mitofusin-2 maintains mitochondrial structure and contributes to stress-induced permeability transition in cardiac myocytes

    Mol. Cell. Biol.

    (2011)
  • Cited by (59)

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text