Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Cell Mol Med. 2019 Mar; 23(3): 1671–1677.
Published online 2019 Jan 16. doi: 10.1111/jcmm.14129
PMCID: PMC6378174
PMID: 30648807

Molecular biomarkers in cardiac hypertrophy

Abstract

Cardiac hypertrophy is characterized by an increase in myocyte size in the absence of cell division. This condition is thought to be an adaptive response to cardiac wall stress resulting from the enhanced cardiac afterload. The pathogenesis of heart dysfunction, which is one of the primary causes of morbidity and mortality in elderly people, is often associated with myocardial remodelling caused by cardiac hypertrophy. In order to well understand the potential mechanisms, we described the molecules involved in the development and progression of myocardial hypertrophy. Increasing evidence has indicated that micro‐RNAs are involved in the pathogenesis of cardiac hypertrophy. In addition, molecular biomarkers including vascular endothelial growth factor B, NAD‐dependent deacetylase sirtuin‐3, growth/differentiation factor 15 and glycoprotein 130, also play important roles in the development of myocardial hypertrophy. Knowing the regulatory mechanisms of these biomarkers in the heart may help identify new molecular targets for the treatment of cardiac hypertrophy.

Keywords: cardiac hypertrophy, micro‐RNAs, molecular biomarkers

1. INTRODUCTION

Myocardial hypertrophy is characterized by the thickening of heart muscles without an obvious cause and found to be involved in several pathological conditions, including hypertension, vascular disease and chronic heart failure.1 Myocardial hypertrophy was first described by Donald Teare in 1958.2, 3 The results of epidemiological studies indicate that the increased incidence and prevalence of heart dysfunction is one of the primary causes of morbidity and mortality in elderly people. The pathogenesis of heart dysfunction is multifactorial and often associated with cardiac remodelling as a result of cardiac myocyte hypertrophy.4, 5, 6 Myocardial hypertrophy is a response to pressure or volume overload whereas chronic left ventricle hypertrophy is primarily associated with chronic heart dysfunction. In addition, myocardial hypertrophy is thought to be a maladaptive process that leads to the fatal gene program and pro‐hypertrophic signalling pathways.7, 8, 9 In the adult heart, the progression of myocardial hypertrophy follows the signals which are stimulated on the cell surface and then transmitted through channels or receptors.10 Therefore, we describe molecules involved in the development and progression of myocardial hypertrophy to well understand underlying mechanisms.

2. MICRO‐RNAS

Micro‐RNAs are non‐coding post‐transcriptional regulatory RNAs that play a key role in regulating mRNA expression and heart diseases.11 Recent studies indicated that changes in the expression levels of micro‐RNAs may contribute to the development of cardiac hypertrophy.12

2.1. MiR‐96

MiR‐96 is a micro‐RNA involved in many diseases caused by impaired cell proliferation.13, 14, 15, 16 It has been confirmed that the mammalian target of rapamycin (mTOR) signalling pathway promotes the onset and progression of cardiac growth.17, 18, 19, 20MiR‐96 is negatively correlated with mTOR and may prevent myocardial hypertrophy by inhibiting mTOR.20 On the other hand, miR‐96 was found to inhibit cardiac hypertrophy by targeting growth factor receptor‐bound 2 which is a negative regulator of cardiac hypertrophy.21 Whether it may offer a new therapeutic strategy for cardiac hypertrophy is still controversial.

2.2. MiR‐30

Several studies have demonstrated that the miR‐30 family micro‐RNAs are involved in the development and progression of tumours and other diseases, including those in the circulatory, genital, respiratory, nervous, alimentary and genital systems. MiR‐30 members play a vital role in cell differentiation, cellular senescence, adipogenesis and drug metabolism.22, 23, 24, 25, 26, 27, 28, 29, 30 Duister et al. found that miR‐30 could trigger myocardial matrix remodelling by regulating the connective tissue growth factor, and the expression of miR‐30 was significantly decreased in mice with hypertrophic cardiomyopathy.31 Reversely associated with p53 up‐regulation, miR‐30 family members are found to inhibit mitochondrial fission and the consequent cardiac hypertrophy related apoptosis.32 Furthermore, cardiac hypertrophy due to chronic alcohol intake in mice was correlated with a lower expression of miR‐30a.33 Another study demonstrated that the expression of beclin‐1 was up‐regulated in cardiomyocytes treated with miR‐30a inhibitor, and the up‐regulation was reversed in cardiomyocytes treated with miR‐30a mimic.34 This study further suggested that the activation of autophagy was enhanced by treating angiotensin II (Ang II)‐induced hypertrophic cardiomyocytes with miR‐30a inhibitor, and this activation was reversed in cells treated with miR‐30a mimic. These results suggest that beclin‐1 can be bound to miR‐30a as a target gene for miR‐30a. Therefore, miR‐30a is a potential diagnostic and therapeutic marker for hypertrophic cardiomyopathy.

2.3. MiR‐34

In mammals, the miR‐34 miRNA precursor can produce three major mature miRNAs. In contrast to other members of this family, the miR‐34 was identified in silico and later confirmed by experiment. In the cell cytoplasm, the precursor miRNA stem‐loop is excised, and the primary miR‐34 mature sequence is removed from the 5′ arm of the hairpin.35, 36, 37 MiR‐34a is a multifunctional regulatory factor that participates in apoptosis,38 cell senescence,39 cell proliferation40 and cell division41 by enhancing or reducing the expression of its target genes. Studies demonstrated that miR‐34a is a component of the p53 tumour suppressor network, suggesting that this mRNA participates in cancer development and progression.42 The expression of miR‐34a varies according to the pathological condition. In myocardial infarction43 and ageing heart,44 the expression of miR‐34a is increased, whereas in most cancers, including bladder and lung cancer, miR‐34a expression is usually decreased.45, 46 Moreover, as a result of transverse aortic constriction, pressure overload leads to changes in miR‐34a expression in different pathological stages of cardiac remodelling,47 such that the expression is decreased in the hypertrophic stage of myocardial remodelling but increased in the heart failure stage. Another study found that miR‐34a extended Caenorhabditis elegans lifespan by inhibiting the activation of ATG9A‐mediated autophagy.48 It is known that Ang II can regulate cardiac hypertrophy and cardiomyocyte autophagy.49 MiR‐34a also can regulate Ang II‐induced cardiomyocyte hypertrophy by directly inhibiting ATG9A expression and autophagic activity.47 It has been found that therapeutic inhibition of the miR‐34 family attenuates pathological cardiac remodelling and improves heart function in mice.50 Therefore, the results of these studies may help identify prospective therapeutic targets for controlling cardiac hypertrophy.

2.4. MiR‐181

The miR‐181 miRNA precursor is a small non‐coding RNA molecule that can generate four mature products: miR‐181a, miR‐181b, miR‐181c and miR‐181d. These products can regulate post‐transcriptional gene expression by binding to target mRNAs. MiR‐181 has been found in a large number of species, including humans, zebrafish and rats. Human miR‐181 has been found in bone marrow, retina and vascular development.51, 53 Furthermore, the expression of miR‐181b was up‐regulated in the blood of patients with myocardial hypertrophy, suggesting that miR‐181b might play a role in both disease pathology and progression.54 Myocardial hypertrophy triggered by stimulation of primary myocardial cells with PE presented a negative correlation between miR‐181b and PKG 1.54 The results suggest that miR‐181b is a promising molecular marker for the clinical diagnosis and treatment of cardiac hypertrophy.

2.5. Other micro‐RNAs

MiR‐378 inhibited caspase‐3 expression in cardiomyocytes and attenuated ischaemic injury, and miR‐199a might be a potential therapeutic target for cardiac hypertrophy or heart failure. It has demonstrated that miR‐185 can suppress the progression of cardiac hypertrophy by reducing ET‐1‐induced hypertrophic responses55 whereas miR‐19a/b positively regulates cardiac hypertrophy by enhancing these responses.56 Lee et al. found that miR‐374 inhibited the cardiac hypertrophy regression pathway by targeting vascular endothelial growth factor receptor 1 (VEGFR‐1) and PKG‐1.57 It is also reported that miR‐9 and miR‐98 were closely related to the development of myocardial hypertrophy.58 Furthermore, the decrease in miR‐133 might reduce endothelin‐1‐ and norepinephrine‐induced myocardial hypertrophy.59

3. VEGF‐B

VEGF‐B is a secretory protein from the VEGF family. VEGF‐A is the best known member of this family, and other members include VEGF‐C and PIGF.60, 61 In humans, VEGF‐B is highly expressed in metabolically active tissues, including fat, heart and skeletal muscle. VEGF‐B has two isoforms produced by alternative splicing, VEGF‐B167 and VEGF‐B186, and both isoforms bind to VEGFR‐1 and neuropilin‐1.62, 63 In contrast to other members of the family, VEGF‐B is not a major angiogenesis‐inducing factor; however, it can enhance neovascularization under pathological conditions.63 It has been reported that VEGF‐B helps maintain newly formed blood vessels during pathological conditions.64 Furthermore, VEGF‐B regulates the uptake and transport of fatty acids in the endothelium of heart and skeletal muscle.65, 66 Karpanen et al. found that up‐regulating the expression of VEGF‐B in mouse heart changed lipid metabolism in myocardial cells and led to myocardial hypertrophy.67 Therefore, VEGF‐B is a potential target for the diagnosis and treatment of myocardial hypertrophy.

4. SIRT3

NAD‐dependent deacetylase sirtuin‐3 (SIRT3) is a member of the mammalian sirtuin family.68 Evidence indicates that SIRT3, as a soluble protein, is located in the mitochondrial matrix and contains a mitochondrial processing peptide at the N‐terminus. Up‐regulating the expression of SIRT3 in vitro inhibits the production of reactive oxygen species and promotes respiration. In white and brown adipose tissue, fasting increases the expression of SIRT3, and the increased expression of SIRT3 in brown adipocytes enhances the expression of UCP1 and PGC‐1α, demonstrating that SIRT3 has a role in adaptive thermogenesis in brown adipose tissue. Furthermore, SIRT3 reduces cardiac hypertrophy by decreasing ROS production.69, 70, 71 SIRT3 augments Foxo3a‐dependent antioxidant defense and further blocks the cardiac hypertrophic response.72

5. GDF15

The growth/differentiation factor 15 (GDF15) was first identified as macrophage inhibitory cytokine‐1.73 It is a member of the transforming growth factor‐beta superfamily. The expression of GDF‐15 in humans is low in most organs and increased by injury to different organs, including heart, kidney, liver and lung.74, 75, 76 GDF‐15 regulates inflammatory pathways and participates in several biological processes, including cellular repair and regulation of apoptosis and cell growth, as well as in cardiovascular and neoplastic disorders.74, 77, 78 GDF‐15 is a powerful prognostic marker in patients with heart disease and cancer.79 Recently, a growing number of studies have focused on the association between GDF‐15 and cardiovascular diseases. It has been proven that GDF‐15 is a good prognostic marker for coronary heart disease and heart failure,80, 81, 82, 83, 84, 85, 86, 87, 88, 89 and participates in cardiac remodelling caused by primary hypertension, hypertrophic cardiomyopathy and ischaemic heart diseases.89, 90, 91, 92, 93 Furthermore, the increased expression of GDF‐15 inhibits norepinephrine‐induced myocardial hypertrophy by decreasing EGF receptor transactivation after norepinephrine stimulation.93 These results indicate that GDF‐15 may be a new target for treating myocardial hypertrophy.

6. GP130

The transmembrane protein glycoprotein 130 (Gp130) is one of the first cytokine receptors to be identified. Gp130 is a signal‐transducing receptor element that binds to the interleukin 6 receptor. Moreover, this molecule is present in several receptors, including IL‐6, IL‐11, ciliary neurotrophic factor and leukaemia inhibitory factor receptors.94, 95, 96 Gp130 is strongly expressed in almost all tissues.96 Nonetheless, the physiological functions of gp130 are not completed explained. A previous study has shown that gp130 has a physiological role in cardiomyocyte regulation, whereas a pathological consequence leading to cardiac hypertrophy will happen after being overstimulated of gp130.97

7. CAMK II

Ca2+/calmodulin‐dependent protein kinase II (CaMKII) is a serine/threonine‐specific protein kinase regulated by the Ca2+/calmodulin complex. CaMKII participates in many signalling pathways and is considered a key mediator of learning and memory.98 In addition, CaMKII contributes to Ca2+ reuptake and homeostasis in cardiomyocytes,99 CD8 T‐cell activation,100 positive T‐cell selection101 and epithelial transport of chloride.102 CaMKII consists of four different isoforms: CaMKIIα, CaMKIIβ, CaMKIIδ and CaMKIIγ. CaMKIIδ is the main cardiac CaMKII isoform, and CaMKIIγ is also expressed in the heart. The expression and activity of CaMKII are enhanced in patients having heart failure.103, 104 Furthermore, the transgenic overexpression of CaMKII induced cardiac hypertrophy and dilated cardiomyopathy.105, 106 Structural heart disease may be prevented by CaMKII inhibitors.107, 108 Moreover, cardiac remodelling was attenuated in mice with global deletion of CaMKIIδ.109, 110 Backs et al. demonstrated that CaMKII could inhibit cardiac hypertrophy via crosstalk with calcineurin.110 Calcineurin has no function in maladaptive cardiac remodelling in the absence of CaMKII signals.111

8. CIC‐3

Chloride channel‐3 (ClC‐3) is a member of the ClC gene family and has been suggested to be a molecular candidate of native volume‐sensitive outwardly rectifying anion channels (VSOAC) in certain mammalian cell types, such as vascular smooth muscle cells and cardiac myocytes.112, 113 In ClC‐3 global knockout mice, the rest of VSOAC in cardiac myocytes caused extensive compensatory changes and altered properties in membrane protein expression.114 It is found that inactivation of ClC‐3 gene produced myocardial hypertrophy and heart failure.114 Furthermore, ClC‐3 is a key molecular of native VSOAC in mammalian heart and plays a crucial role in preventing myocardial hypertrophy.114

9. OTHER MOLECULES

The expression of adhesion molecules including CD11a, CD11b and CD11c was increased in rats with myocardial hypertrophy.115 Walsh reported that the inhibition of stress‐induced activin A/Smad2 signalling triggered the expression of follistatin‐like in hypertrophied cardiac myocytes.116 Follistatin‐like 3 is a stress‐induced regulator of cardiac hypertrophy and may regulate myocyte size via Smad signalling.116 Moreover, the expression of N‐cadherin was up‐regulated in myocardial tissues in rats. Although these findings are based on animal experimentation, it provides additional motivation for researchers to explore the functions of these biomarkers in regulating myocardial hypertrophy in human beings, either in modulating physiological environment or developing molecular target drugs.

10. CONCLUSIONS

Myocardial hypertrophy is adaptive response to cardiac wall stress resulting from the enhanced cardiac load. The main feature of myocardial hypertrophy is the embryonic genes up‐regulation, protein synthesis increases and cell volume expansion. The predominant histopathological characteristics are expanded cell space, increased intercellular muscle fibres and myocardial fibrosis and dysfunction. In this review, we list several micro‐RNAs and other molecular biomarkers involved in the pathogenesis of cardiac hypertrophy (Table 1). Knowing the regulatory mechanisms of these biomarkers may help identify new molecular targets for the treatment of cardiac hypertrophy.

Table 1

Molecular biomarkers involved in cardiac hypertrophy

BiomarkersCharacteristics
micro‐RNAs
MiR‐96Inhibit mTOR1 and reduce growth factor receptor‐bound 2 expression20, 21
MiR‐30Inhibit cardiac hypertrophy related apoptosis32
MiR‐34Up‐regulated in the heart in response to stress; improve cardiac function50
MiR‐181Up‐regulated in myocardial hypertrophy54
VEGF‐BUp‐regulated in lipid metabolism in myocardial cells65, 66; lead to myocardial hypertrophy67
SIRT3Block cardiac hypertrophic response by augmenting Foxo3a‐dependent antioxidant defense72
GDF15Inhibit norepinephrine‐induced myocardial hypertrophy93
GP130Lead to cardiac hypertrophy by activating its expression97
CaMK IIIncreased in hypertrophied myocardium and related to cardiac hypertrophy105, 106, 107, 108, 109, 110, 111
CIC‐3Associated with myocardial hypertrophy and heart failure114

CaMK II, calmodulin‐dependent protein kinase II; ClC‐3, chloride channel‐3; GDF15, growth/differentiation factor 15; GP130, glycoprotein 130; SIRT3, NAD‐dependent deacetylase sirtuin‐3; VEGF‐B, vascular endothelial growth factor B; mTOR1, mammalian target of rapamycin 1.

CONFLICT OF INTEREST

The authors declare that there are no conflicts of interest.

ACKNOWLEDGEMENT

This study was financially supported by the National Natural Science Foundation of China (No. 81770370).

Notes

Zhu L, Li C, Liu Q, Xu W, Zhou X. Molecular biomarkers in cardiac hypertrophy. J Cell Mol Med. 2019;23:1671‐1677. 10.1111/jcmm.14129 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Liu Zhu and Chao Li equally contributed to this work.

REFERENCES

1. Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol. 2016;97:245‐262. [PubMed] [Google Scholar]
2. Teare D. Asymmetrical hypertrophy of the heart in young adults. Br Heart J. 1958;20:1‐8. [PMC free article] [PubMed] [Google Scholar]
3. McKenna WJ, Sen‐Chowdhry S. From Teare to the present day: a fifty year odyssey in hypertrophic cardiomyopathy, a paradigm for the logic of the discovery process. Rev Esp Cardiol. 2008;61:1239‐1244. [PubMed] [Google Scholar]
4. Ashrafian H, Frenneaux MP, Opie LH. Metabolic mechanisms in heart failure. Circulation. 2007;116:434‐448. [PubMed] [Google Scholar]
5. Yan L, Wei X, Tang QZ, et al. Cardiac‐specific mindin overexpression attenuates cardiac hypertrophy via blocking AKT/GSK3beta and TGF‐beta1‐Smad signalling. Cardiovasc Res. 2011;92:85‐94. [PMC free article] [PubMed] [Google Scholar]
6. Frey N, Katus HA, Olson EN, Hill JA. Hypertrophy of the heart: a new therapeutic target? Circulation. 2004;109:1580‐1589. [PubMed] [Google Scholar]
7. Manyari DE. Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med. 1990;323:1706‐1707. [PubMed] [Google Scholar]
8. Satoh M, Ogita H, Takeshita K, Mukai Y, Kwiatkowski DJ, Liao JK. Requirement of Rac1 in the development of cardiac hypertrophy. Proc Natl Acad Sci USA. 2006;103:7432‐7437. [PMC free article] [PubMed] [Google Scholar]
9. Olson EN, Schneider MD. Sizing up the heart: development redux in disease. Genes Dev. 2003;17:1937‐1956. [PubMed] [Google Scholar]
10. Molkentin JD, Dorn GW 2nd. Cytoplasmic signaling pathways that regulate cardiac hypertrophy. Annu Rev Physiol. 2001;63:391‐426. [PubMed] [Google Scholar]
11. Yamada Y, Enokida H, Kojima S, et al. MiR‐96 and miR‐183 detection in urine serve as potential tumor markers of urothelial carcinoma: correlation with stage and grade, and comparison with urinary cytology. Cancer Sci. 2011;102:522‐529. [PubMed] [Google Scholar]
12. Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP. Vertebrate microRNA genes. Science. 2003;299:1540. [PubMed] [Google Scholar]
13. Xu XM, Qian JC, Deng ZL, et al. Expression of miR‐21, miR‐31, miR‐96 and miR‐135b is correlated with the clinical parameters of colorectal cancer. Oncol Lett. 2012;4:339‐345. [PMC free article] [PubMed] [Google Scholar]
14. Liu Y, Han Y, Zhang H, et al. Synthetic miRNA‐mowers targeting miR‐183‐96‐182 cluster or miR‐210 inhibit growth and migration and induce apoptosis in bladder cancer cells. PLoS ONE. 2012;7:e52280. [PMC free article] [PubMed] [Google Scholar]
15. Zhang W, Qian P, Zhang X, et al. Autocrine/paracrine human growth hormone‐stimulated microRNA 96‐182‐183 cluster promotes epithelial‐mesenchymal transition and invasion in breast cancer. J Biol Chem. 2015;290:13812‐13829. [PMC free article] [PubMed] [Google Scholar]
16. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21‐35. [PMC free article] [PubMed] [Google Scholar]
17. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149:274‐293. [PMC free article] [PubMed] [Google Scholar]
18. Liu Y, Yang F, Ma W, Sun Q. Metformin inhibits proliferation and proinflammatory cytokines of human keratinocytes in vitro via mTOR‐signaling pathway. Pharm Biol. 2016;54:1173‐1178. [PubMed] [Google Scholar]
19. Huang TQ, Zou MX, Pasek DA, Meissner G. mTOR signaling in mice with dysfunctional cardiac ryanodine receptor ion channel. J Receptor Ligand Channel Res. 2015;8:43‐51. [PMC free article] [PubMed] [Google Scholar]
20. Sun X, Zhang C. MicroRNA‐96 promotes myocardial hypertrophy by targeting mTOR. Int J Clin Exp Pathol. 2015;8:14500‐14506. [PMC free article] [PubMed] [Google Scholar]
21. Xia Y, Sheng J, Liang GY, Liu DX, Tang Q, Cheng AP. miR‐96 inhibits cardiac hypertrophy by targeting growth factor receptor‐bound 2. Genet Mol Res. 2015;14:18958‐18964. [PubMed] [Google Scholar]
22. Mellios N, Galdzicka M, Ginns E, et al. Gender‐specific reduction of estrogen‐sensitive small RNA, miR‐30b, in subjects with schizophrenia. Schizophr Bull. 2012;38:433‐443. [PMC free article] [PubMed] [Google Scholar]
23. Zaragosi LE, Wdziekonski B, Brigand KL, et al. Small RNA sequencing reveals miR‐642a‐3p as a novel adipocyte‐specific microRNA and miR‐30 as a key regulator of human adipogenesis. Genome Biol. 2011;12:R64. [PMC free article] [PubMed] [Google Scholar]
24. Chartoumpekis DV, Zaravinos A, Ziros PG, et al. Differential expression of microRNAs in adipose tissue after long‐term high‐fat diet‐induced obesity in mice. PLoS ONE. 2012;7:e34872. [PMC free article] [PubMed] [Google Scholar]
25. Martinez I, Cazalla D, Almstead LL, Steitz JA, DiMaio D. miR‐29 and miR‐30 regulate B‐Myb expression during cellular senescence. Proc Natl Acad Sci USA. 2011;108:522‐527. [PMC free article] [PubMed] [Google Scholar]
26. Caruso P, MacLean MR, Khanin R, et al. Dynamic changes in lung microRNA profiles during the development of pulmonary hypertension due to chronic hypoxia and monocrotaline. Arterioscler Thromb Vasc Biol. 2010;30:716‐723. [PubMed] [Google Scholar]
27. Lin J, Lwin T, Zhao JJ, et al. Follicular dendritic cell‐induced microRNA‐mediated upregulation of PRDM1 and downregulation of BCL‐6 in non‐Hodgkin's B‐cell lymphomas. Leukemia. 2011;25:145‐152. [PMC free article] [PubMed] [Google Scholar]
28. Yu Y, Yang L, Zhao M, et al. Targeting microRNA‐30a‐mediated autophagy enhances imatinib activity against human chronic myeloid leukemia cells. Leukemia. 2012;26:1752‐1760. [PubMed] [Google Scholar]
29. Zhong X, Li N, Liang S, Huang Q, Coukos G, Zhang L. Identification of microRNAs regulating reprogramming factor LIN28 in embryonic stem cells and cancer cells. J Biol Chem. 2010;285:41961‐41971. [PMC free article] [PubMed] [Google Scholar]
30. Wu T, Zhou H, Hong Y, Li J, Jiang X, Huang H. miR‐30 family members negatively regulate osteoblast differentiation. J Biol Chem. 2012;287:7503‐7511. [PMC free article] [PubMed] [Google Scholar]
31. Duisters RF, Tijsen AJ, Schroen B, et al. miR‐133 and miR‐30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling. Circ Res. 2009;104:170‐178, 176 p. following 178. [PubMed] [Google Scholar]
32. Li JC, Donath S, Li YR, Qin D, Prabhakar BS, Li PF. miR‐30 regulates mitochondrial fission through targeting p53 and the dynamin‐related protein‐1 pathway. PLoS Genet. 2010;6:e1000795. [PMC free article] [PubMed] [Google Scholar]
33. Guo R, Hu N, Kandadi MR, Ren J. Facilitated ethanol metabolism promotes cardiomyocyte contractile dysfunction through autophagy in murine hearts. Autophagy. 2012;8:593‐608. [PMC free article] [PubMed] [Google Scholar]
34. Pan W, Zhong Y, Cheng C, et al. MiR‐30‐regulated autophagy mediates angiotensin II‐induced myocardial hypertrophy. PLoS ONE. 2013;8:e53950. [PMC free article] [PubMed] [Google Scholar]
35. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007;100:416‐424. [PubMed] [Google Scholar]
36. Dostie J, Mourelatos Z, Yang M, Sharma A, Dreyfuss G. Numerous microRNPs in neuronal cells containing novel microRNAs. RNA. 2003;9:180‐186. [PMC free article] [PubMed] [Google Scholar]
37. Landgraf P, Rusu M, Sheridan R, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129:1401‐1414. [PMC free article] [PubMed] [Google Scholar]
38. He L, He X, Lim LP, et al. A microRNA component of the p53 tumour suppressor network. Nature. 2007;447:1130‐1134. [PMC free article] [PubMed] [Google Scholar]
39. Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. Tumor‐suppressive miR‐34a induces senescence‐like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA. 2007;104:15472‐15477. [PMC free article] [PubMed] [Google Scholar]
40. Tivnan A, Tracey L, Buckley PG, Alcock LC, Davidoff AM, Stallings RL. MicroRNA‐34a is a potent tumor suppressor molecule in vivo in neuroblastoma. BMC Cancer. 2011;11:33. [PMC free article] [PubMed] [Google Scholar]
41. Fujita Y, Kojima K, Hamada N, et al. Effects of miR‐34a on cell growth and chemoresistance in prostate cancer PC3 cells. Biochem Biophys Res Comm. 2008;377:114‐119. [PubMed] [Google Scholar]
42. Lui WO, Pourmand N, Patterson BK, Fire A. Patterns of known and novel small RNAs in human cervical cancer. Can Res. 2007;67:6031‐6043. [PubMed] [Google Scholar]
43. Cheng Y, Ji R, Yue J, et al. MicroRNAs are aberrantly expressed in hypertrophic heart: do they play a role in cardiac hypertrophy? Am J Pathol. 2007;170:1831‐1840. [PMC free article] [PubMed] [Google Scholar]
44. Boon RA, Iekushi K, Lechner S, et al. MicroRNA‐34a regulates cardiac ageing and function. Nature. 2013;495:107‐110. [PubMed] [Google Scholar]
45. Basak SK, Veena MS, Oh S, et al. The CD44(high) tumorigenic subsets in lung cancer biospecimens are enriched for low miR‐34a expression. PLoS ONE. 2013;8:e73195. [PMC free article] [PubMed] [Google Scholar]
46. Wang W, Li T, Han G, Li Y, Shi LH, Li H. Expression and role of miR‐34a in bladder cancer. Indian J Biochem Biophys. 2013;50:87‐92. [PubMed] [Google Scholar]
47. Huang J, Sun W, Huang H, et al. miR‐34a modulates angiotensin II‐induced myocardial hypertrophy by direct inhibition of ATG9A expression and autophagic activity. PLoS ONE. 2014;9:e94382. [PMC free article] [PubMed] [Google Scholar]
48. Yang J, Chen D, He Y, et al. MiR‐34 modulates Caenorhabditis elegans lifespan via repressing the autophagy gene atg9. Age. 2013;35:11‐22. [PMC free article] [PubMed] [Google Scholar]
49. Porrello ER, Delbridge LM. Cardiomyocyte autophagy is regulated by angiotensin II type 1 and type 2 receptors. Autophagy. 2009;5:1215‐1216. [PubMed] [Google Scholar]
50. Bernardo BC, Gao XM, Winbanks CE, et al. Therapeutic inhibition of the miR‐34 family attenuates pathological cardiac remodeling and improves heart function. Proc Natl Acad Sci USA. 2012;109:17615‐17620. [PMC free article] [PubMed] [Google Scholar]
51. Ryan DG, Oliveira‐Fernandes M, Lavker RM, et al. MicroRNAs of the mammalian eye display distinct and overlapping tissue specificity. Mol Vis. 2006;12:1175‐1184. [PubMed] [Google Scholar]
52. Chen CZ, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage differentiation. Science. 2004;303:83‐86. [PubMed] [Google Scholar]
53. Domigan CK, Iruela‐Arispe ML. Recent advances in vascular development. Curr Opin Hematol. 2012;19:176‐183. [PMC free article] [PubMed] [Google Scholar]
54. Zhong W, Yang J, Cao Q, Huan X. Association between miR‐181b and PKG 1 in myocardial hypertrophy and its clinical implications. Exp Ther Med. 2015;10:857‐862. [PMC free article] [PubMed] [Google Scholar]
55. Kim JO, Song DW, Kwon EJ, et al. miR‐185 plays an anti‐hypertrophic role in the heart via multiple targets in the calcium‐signaling pathways. PLoS ONE. 2015;10:e0122509. [PMC free article] [PubMed] [Google Scholar]
56. Song DW, Ryu JY, Kim JO, Kwon EJ, Kim DH. The miR‐19a/b family positively regulates cardiomyocyte hypertrophy by targeting atrogin‐1 and MuRF‐1. Biochem J. 2014;457:151‐162. [PubMed] [Google Scholar]
57. Lee JS, Song DW, Park JH, Kim JO, Cho C, Kim DH. miR‐374 promotes myocardial hypertrophy by negatively regulating vascular endothelial growth factor receptor‐1 signaling. BMB Rep. 2017;50:208‐213. [PMC free article] [PubMed] [Google Scholar]
58. Harada M, Luo X, Murohara T, Yang B, Dobrev D, Nattel S. MicroRNA regulation and cardiac calcium signaling: role in cardiac disease and therapeutic potential. Circ Res. 2014;114:689‐705. [PubMed] [Google Scholar]
59. Katta A, Thulluri S, Manne ND, et al. Overload induced heat shock proteins (HSPs), MAPK and miRNA (miR‐1 and miR133a) response in insulin‐resistant skeletal muscle. Cell Physiol Biochem. 2013;31:219‐229. [PubMed] [Google Scholar]
60. Karpanen T, Alitalo K. Molecular biology and pathology of lymphangiogenesis. Annu Rev Pathol. 2008;3:367‐397. [PubMed] [Google Scholar]
61. Aase K, Lymboussaki A, Kaipainen A, Olofsson B, Alitalo K, Eriksson U. Localization of VEGF‐B in the mouse embryo suggests a paracrine role of the growth factor in the developing vasculature. Dev Dyn. 1999;215:12‐25. [PubMed] [Google Scholar]
62. Olofsson B, Korpelainen E, Pepper MS, et al. Vascular endothelial growth factor B (VEGF‐B) binds to VEGF receptor‐1 and regulates plasminogen activator activity in endothelial cells. Proc Natl Acad Sci USA. 1998;95:11709‐11714. [PMC free article] [PubMed] [Google Scholar]
63. Makinen T, Olofsson B, Karpanen T, et al. Differential binding of vascular endothelial growth factor B splice and proteolytic isoforms to neuropilin‐1. J Biol Chem. 1999;274:21217‐21222. [PubMed] [Google Scholar]
64. Zhang F, Tang Z, Hou X, et al. VEGF‐B is dispensable for blood vessel growth but critical for their survival, and VEGF‐B targeting inhibits pathological angiogenesis. Proc Natl Acad Sci USA. 2009;106:6152‐6157. [PMC free article] [PubMed] [Google Scholar]
65. Muoio DM. Metabolism and vascular fatty acid transport. N Engl J Med. 2010;363:291‐293. [PubMed] [Google Scholar]
66. Hagberg CE, Mehlem A, Falkevall A, et al. Targeting VEGF‐B as a novel treatment for insulin resistance and type 2 diabetes. Nature. 2012;490:426‐430. [PubMed] [Google Scholar]
67. Karpanen T, Bry M, Ollila HM, et al. Overexpression of vascular endothelial growth factor‐B in mouse heart alters cardiac lipid metabolism and induces myocardial hypertrophy. Circ Res. 2008;103:1018‐1026. [PMC free article] [PubMed] [Google Scholar]
68. Schwer B, North BJ, Frye RA, Ott M, Verdin E. The human silent information regulator (Sir)2 homologue hSIRT3 is a mitochondrial nicotinamide adenine dinucleotide‐dependent deacetylase. J Cell Biol. 2002;158:647‐657. [PMC free article] [PubMed] [Google Scholar]
69. Lang D. Cardiac hypertrophy and oxidative stress: a leap of faith or stark reality? Heart. 2002;87:316‐317. [PMC free article] [PubMed] [Google Scholar]
70. Zweier JL, Talukder MA. The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res. 2006;70:181‐190. [PubMed] [Google Scholar]
71. Li J, Chen T, Xiao M, et al. Mouse SIRT3 promotes autophagy in AngII‐induced myocardial hypertrophy through the deacetylation of FoxO1. Oncotarget. 2016;7:86648‐86659. [PMC free article] [PubMed] [Google Scholar]
72. Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP. SIRT3 blocks the cardiac hypertrophic response by augmenting Foxo3a‐dependent antioxidant defense mechanisms in mice. J Clin Invest. 2009;119:2758‐2771. [PMC free article] [PubMed] [Google Scholar]
73. Zimmers TA, Jin X, Hsiao EC, McGrath SA, Esquela AF, Koniaris LG. Growth differentiation factor‐15/macrophage inhibitory cytokine‐1 induction after kidney and lung injury. Shock. 2005;23:543‐548. [PubMed] [Google Scholar]
74. Hsiao EC, Koniaris LG, Zimmers‐Koniaris T, Sebald SM, Huynh TV, Lee SJ. Characterization of growth‐differentiation factor 15, a transforming growth factor beta superfamily member induced following liver injury. Mol Cell Biol. 2000;20:3742‐3751. [PMC free article] [PubMed] [Google Scholar]
75. Ago T, Sadoshima J. GDF15, a cardioprotective TGF‐beta superfamily protein. Circ Res. 2006;98:294‐297. [PubMed] [Google Scholar]
76. Wollert KC, Kempf T, Lagerqvist B, et al. Growth differentiation factor 15 for risk stratification and selection of an invasive treatment strategy in non ST‐elevation acute coronary syndrome. Circulation. 2007;116:1540‐1548. [PubMed] [Google Scholar]
77. Kempf T, Eden M, Strelau J, et al. The transforming growth factor‐beta superfamily member growth‐differentiation factor‐15 protects the heart from ischemia/reperfusion injury. Circ Res. 2006;98:351‐360. [PubMed] [Google Scholar]
78. Wallentin L, Zethelius B, Berglund L, et al. GDF‐15 for prognostication of cardiovascular and cancer morbidity and mortality in men. PLoS ONE. 2013;8:e78797. [PMC free article] [PubMed] [Google Scholar]
79. Wollert KC, Kempf T, Peter T, et al. Prognostic value of growth‐differentiation factor‐15 in patients with non‐ST‐elevation acute coronary syndrome. Circulation. 2007;115:962‐971. [PubMed] [Google Scholar]
80. Kempf T, von Haehling S, Peter T, et al. Prognostic utility of growth differentiation factor‐15 in patients with chronic heart failure. J Am Coll Cardiol. 2007;50:1054‐1060. [PubMed] [Google Scholar]
81. Kempf T, Bjorklund E, Olofsson S, et al. Growth‐differentiation factor‐15 improves risk stratification in ST‐segment elevation myocardial infarction. Eur Heart J. 2007;28:2858‐2865. [PubMed] [Google Scholar]
82. Khan SQ, Ng K, Dhillon O, et al. Growth differentiation factor‐15 as a prognostic marker in patients with acute myocardial infarction. Eur Heart J. 2009;30:1057‐1065. [PubMed] [Google Scholar]
83. Anand IS, Kempf T, Rector TS, et al. Serial measurement of growth‐differentiation factor‐15 in heart failure: relation to disease severity and prognosis in the Valsartan Heart Failure Trial. Circulation. 2010;122:1387‐1395. [PubMed] [Google Scholar]
84. Eggers KM, Kempf T, Lagerqvist B, et al. Growth‐differentiation factor‐15 for long‐term risk prediction in patients stabilized after an episode of non‐ST‐segment‐elevation acute coronary syndrome. Circ Cardiovasc Gene. 2010;3:88‐96. [PubMed] [Google Scholar]
85. Kempf T, Horn‐Wichmann R, Brabant G, et al. Circulating concentrations of growth‐differentiation factor 15 in apparently healthy elderly individuals and patients with chronic heart failure as assessed by a new immunoradiometric sandwich assay. Clin Chem. 2007;53:284‐291. [PubMed] [Google Scholar]
86. Eggers KM, Kempf T, Lind L, et al. Relations of growth‐differentiation factor‐15 to biomarkers reflecting vascular pathologies in a population‐based sample of elderly subjects. Scand J Clin Lab Invest. 2012;72:45‐51. [PubMed] [Google Scholar]
87. Wang F, Guo Y, Yu H, Zheng L, Mi L, Gao W. Growth differentiation factor 15 in different stages of heart failure: potential screening implications. Biomarkers. 2010;15:671‐676. [PubMed] [Google Scholar]
88. Dominguez‐Rodriguez A, Abreu‐Gonzalez P, Avanzas P. Relation of growth‐differentiation factor 15 to left ventricular remodeling in ST‐segment elevation myocardial infarction. Am J Cardiol. 2011;108:955‐958. [PubMed] [Google Scholar]
89. Lind L, Wallentin L, Kempf T, et al. Growth‐differentiation factor‐15 is an independent marker of cardiovascular dysfunction and disease in the elderly: results from the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) Study. Eur Heart J. 2009;30:2346‐2353. [PubMed] [Google Scholar]
90. Wang X, Yang X, Sun K, et al. The haplotype of the growth‐differentiation factor 15 gene is associated with left ventricular hypertrophy in human essential hypertension. Clin Sci. 2009;118:137‐145. [PubMed] [Google Scholar]
91. Bjornstad JL, Neverdal NO, Vengen OA, et al. Alterations in circulating activin A, GDF‐15, TGF‐beta3 and MMP‐2, ‐3, and ‐9 during one year of left ventricular reverse remodelling in patients operated for severe aortic stenosis. Eur J Heart Fail. 2008;10:1201‐1207. [PubMed] [Google Scholar]
92. Montoro‐Garcia S, Hernandez‐Romero D, Jover E, et al. Growth differentiation factor‐15, a novel biomarker related with disease severity in patients with hypertrophic cardiomyopathy. Eur J Intern Med. 2012;23:169‐174. [PubMed] [Google Scholar]
93. Xu XY, Nie Y, Wang FF, et al. Growth differentiation factor (GDF)‐15 blocks norepinephrine‐induced myocardial hypertrophy via a novel pathway involving inhibition of epidermal growth factor receptor transactivation. J Biol Chem. 2014;289:10084‐10094. [PMC free article] [PubMed] [Google Scholar]
94. Hibi M, Murakami M, Saito M, Hirano T, Taga T, Kishimoto T. Molecular cloning and expression of an IL‐6 signal transducer, gp130. Cell. 1990;63:1149‐1157. [PubMed] [Google Scholar]
95. Davis S, Aldrich TH, Stahl N, et al. LIFR beta and gp130 as heterodimerizing signal transducers of the tripartite CNTF receptor. Science. 1993;260:1805‐1808. [PubMed] [Google Scholar]
96. Saito M, Yoshida K, Hibi M, Taga T, Kishimoto T. Molecular cloning of a murine IL‐6 receptor‐associated signal transducer, gp130, and its regulated expression in vivo. J Immunol. 1992;148:4066‐4071. [PubMed] [Google Scholar]
97. Hirota H, Yoshida K, Kishimoto T, Taga T. Continuous activation of gp130, a signal‐transducing receptor component for interleukin 6‐related cytokines, causes myocardial hypertrophy in mice. Proc Natl Acad Sci USA. 1995;92:4862‐4866. [PMC free article] [PubMed] [Google Scholar]
98. Yamauchi T. Neuronal Ca2+/calmodulin‐dependent protein kinase II–discovery, progress in a quarter of a century, and perspective: implication for learning and memory. Biol Pharm Bull. 2005;28:1342‐1354. [PubMed] [Google Scholar]
99. Anderson ME. Calmodulin kinase signaling in heart: an intriguing candidate target for therapy of myocardial dysfunction and arrhythmias. Pharmacol Ther. 2005;106:39‐55. [PubMed] [Google Scholar]
100. Lin MY, Zal T, Ch'en IL, Gascoigne NR, Hedrick SM. A pivotal role for the multifunctional calcium/calmodulin‐dependent protein kinase II in T cells: from activation to unresponsiveness. J Immunol. 2005;174:5583‐5592. [PubMed] [Google Scholar]
101. McGargill MA, Sharp LL, Bui JD, Hedrick SM, Calbo S. Active Ca2+/calmodulin‐dependent protein kinase II gamma B impairs positive selection of T cells by modulating TCR signaling. J Immunol. 2005;175:656‐664. [PubMed] [Google Scholar]
102. Fahrmann M, Kaufhold MA. Functional partitioning of epithelial protein kinase CaMKII in signal transduction. Biochem Biophys Acta. 2006;1763:101‐109. [PubMed] [Google Scholar]
103. Colomer JM, Mao L, Rockman HA, Means AR. Pressure overload selectively up‐regulates Ca2+/calmodulin‐dependent protein kinase II in vivo. Mol Endocrinol. 2003;17:183‐192. [PubMed] [Google Scholar]
104. Hoch B, Meyer R, Hetzer R, Krause EG, Karczewski P. Identification and expression of delta‐isoforms of the multifunctional Ca2+/calmodulin‐dependent protein kinase in failing and nonfailing human myocardium. Circ Res. 1999;84:713‐721. [PubMed] [Google Scholar]
105. Zhang T, Johnson EN, Gu Y, et al. The cardiac‐specific nuclear delta(B) isoform of Ca2+/calmodulin‐dependent protein kinase II induces hypertrophy and dilated cardiomyopathy associated with increased protein phosphatase 2A activity. J Biol Chem. 2002;277:1261‐1267. [PubMed] [Google Scholar]
106. Zhang T, Maier LS, Dalton ND, et al. The deltaC isoform of CaMKII is activated in cardiac hypertrophy and induces dilated cardiomyopathy and heart failure. Circ Res. 2003;92:912‐919. [PubMed] [Google Scholar]
107. Joiner ML, Koval OM, Li J, et al. CaMKII determines mitochondrial stress responses in heart. Nature. 2012;491:269‐273. [PMC free article] [PubMed] [Google Scholar]
108. Zhang R, Khoo MS, Wu Y, et al. Calmodulin kinase II inhibition protects against structural heart disease. Nat Med. 2005;11:409‐417. [PubMed] [Google Scholar]
109. Ling H, Zhang T, Pereira L, et al. Requirement for Ca2+/calmodulin‐dependent kinase II in the transition from pressure overload‐induced cardiac hypertrophy to heart failure in mice. J Clin Investig. 2009;119:1230‐1240. [PMC free article] [PubMed] [Google Scholar]
110. Backs J, Backs T, Neef S, et al. The delta isoform of CaM kinase II is required for pathological cardiac hypertrophy and remodeling after pressure overload. Proc Natl Acad Sci USA. 2009;106:2342‐2347. [PMC free article] [PubMed] [Google Scholar]
111. Kreusser MM, Lehmann LH, Keranov S, et al. Cardiac CaM Kinase II genes delta and gamma contribute to adverse remodeling but redundantly inhibit calcineurin‐induced myocardial hypertrophy. Circulation. 2014;130:1262‐1273. [PMC free article] [PubMed] [Google Scholar]
112. Yamazaki J, Duan D, Janiak R, Kuenzli K, Horowitz B, Hume JR. Functional and molecular expression of volume‐regulated chloride channels in canine vascular smooth muscle cells. J Physiol. 1998;507:729‐736. [PMC free article] [PubMed] [Google Scholar]
113. Yamamoto‐Mizuma S, Wang GX, Liu LL, et al. Altered properties of volume‐sensitive osmolyte and anion channels (VSOACs) and membrane protein expression in cardiac and smooth muscle myocytes from Clcn3−/− mice. J Physiol. 2004;557:439‐456. [PMC free article] [PubMed] [Google Scholar]
114. Xiong D, Heyman NS, Airey J, et al. Cardiac‐specific, inducible ClC‐3 gene deletion eliminates native volume‐sensitive chloride channels and produces myocardial hypertrophy in adult mice. J Mol Cell Cardiol. 2010;48:211‐219. [PMC free article] [PubMed] [Google Scholar]
115. Mu L, Jing C, Guo Z. Expressions of CD11a, CD11b, and CD11c integrin proteins in rats with myocardial hypertrophy. Iran J Basic Med Sci. 2014;17:874‐878. [PMC free article] [PubMed] [Google Scholar]
116. Shimano M, Ouchi N, Nakamura K, et al. Cardiac myocyte‐specific ablation of follistatin‐like 3 attenuates stress‐induced myocardial hypertrophy. J Biol Chem. 2011;286:9840‐9848. [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

-