Open Access

The diagnostic value of circulating microRNAs in heart failure (Review)

  • Authors:
    • Yao‑Meng Huang
    • Wei‑Wei Li
    • Jun Wu
    • Mei Han
    • Bing‑Hui Li
  • View Affiliations

  • Published online on: January 15, 2019     https://doi.org/10.3892/etm.2019.7177
  • Pages: 1985-2003
  • Copyright: © Huang 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

Heart failure (HF) is a complex clinical syndrome, characterized by inadequate blood perfusion of tissues and organs caused by decreased heart ejection capacity resulting from structural or functional cardiac disorders. HF is the most severe heart condition and it severely compromises human health; thus, its early diagnosis and effective management are crucial. However, given the lack of satisfactory sensitivity and specificity of the currently available biomarkers, the majority of patients with HF are not diagnosed early and do not receive timely treatment. A number of studies have demonstrated that peripheral blood circulating nucleic acids [such as microRNAs (miRs), mRNA and DNA] are important for the diagnosis and monitoring of treatment response in HF. miRs have been attracting increasing attention as promising biomarkers, given their presence in body fluids and relative structural stability under diverse conditions of sampling. The aim of the present review was to analyze the associations between the mechanisms underlying the development of HF and the expression of miRs, and discuss the value of using circulating miRs as diagnostic biomarkers in HF management. In particular, miR‑155, miR‑22 and miR‑133 appear to be promising for the diagnosis, prognosis and management of HF patients.

Introduction

The causes of heart failure (HF) include ischemic cardiomyopathy (ICM) and dilated cardiomyopathy (DCM), hypertension, valvular heart disease, diabetic cardiomyopathy and congenital heart disease (CHD) (1). The pathogenesis of HF is associated with myocardial hypertrophy, fibrosis or necrosis, cardiomyocyte apoptosis, renin-angiotensin-aldosterone system imbalance and collagen changes, as well as several other factors (27).

MicroRNAs (miRs) are small (~22 nucleotides in length), single-strand, non-coding RNA sequences derived from precursors that control gene expression in a variety of physiological and developmental processes, which are involved in post-transcriptional regulation of gene expression (8). miR disorders are associated with a number of human diseases, including diabetes, myocardial infarction and cardiovascular disease, obesity and cancer. Several studies have demonstrated that miRs may affect different aspects of the occurrence and development of HF (914). The association between miRs and HF is discussed in detail below.

Circulating miRs are increasingly recognized as promising biomarkers, given their stability and resistance to endogenous RNase (15); these miRs, to some degree, may also be used as diagnostic biomarkers for angiocardiopathy. In addition, miRNAs and various types of HF have complex relationships, as described below.

Changes and associated mechanisms of miRs in various types of HF

miRs may be involved in several aspects of the occurrence and development of HF, such as cardiomyocyte apoptosis, hypertrophy, fibrosis, inflammation, oxidative damage and hypoxic damage (914), among others. The specific regulatory functions of miRs are indicated in Figs. 1 and 2 and are summarized in Table I (1666).

Figure 2.

Association between miRs and different pathogenic mechanisms of heart failure. Solid lines represent positive regulation and dashed lines represent negative regulation. The nock of the arrow controls the tip of the arrow, for example miR-451 downregulates the LKB1/AMPK pathway, and the LKB1/AMPK pathway negatively regulates the tendency for cardiomyocyte hypertrophy. Therefore, miR-451 promotes myocardial hypertrophy. JUN, Jun proto-oncogene product which is a subunit of the AP-1 transcription; HOXA9, Homeobox A9; UCA1, urothelial carcinoma-associated 1; KLF13, Kruppel-like transcription factor 13; CIAPIN1, cytokine-induced anti-apoptotic molecule; BDNF, brain derived neurotrophic factor; TGFβ-1, transforming growth factor β-1; Bcl-2, B-cell lymphoma-2; APAF-1, apoptotic protease activating factor-1; SGK, Serum and Glucocorticoid Induced Kinase; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; ITGB3, integrin β3; PTEN, phosphatase and tensin homolog deleted on chromosome 10; MCU, mitochondrial calcium uptake; INSR, insulin receptor; IGFR1, Insulin-like growth factor 1 receptor; SIRT4, Sirtuin-4; fos-AP1, Fos-Associated Protein 1; MMP, matrix metalloproteinase; AMPK, adenosine monophosphate-activated protein kinase; LKB1, Liver kinase B1; NF-κB, nuclear factor kappaB; TRAF3, TNF receptor associated factor 3; TNF-α, tumor necrosis factor-α; IL-6, interleukin-6; AP-1, activator protein-1; ALK-5, activin-like kinase 5; PKC, protein kinase C; CAV3, Caveolin 3; ROS, reactive oxygen species; COX, cycloxygenase.

Table I.

Information on different types of miRs.

Table I.

Information on different types of miRs.

Author, yearmiRsRelative gene protein or signaling pathwaysBiological effectsTissue, cells or experimental modelDetection means(Refs.)
Li et al, 2016miR-7a/bSp1, PARP-1 and caspase-3Binding activity of Sp1 may conditionally mediate the repression of miR-7a/b-regulated PARP-1 and caspase-3 expression, miR-7a/b inhibitors effectively upregulated Sp1, PARP-1 and caspase-3 expressionH9c2 cell lineWestern blot, RT-qPCR, luciferase assay and ChIP(16)
Ball et al, 2017miR-21ALDO/SALT, TGFβ-2, IL-1β, MCP-1, Col3a1 and Col1a1miR-21 downregulation attenuated ALDO/SALT-mediated LV inflammatory marker mRNA expression, such as TGFβ-2, IL-1β and MCP-1. miR-21 downregulation exacerbated ALDO/SALT-mediated LV fibrosis marker mRNA expression upregulation, such as Col1a1 and Col3a1Left ventricle of Control, ALDO, SALT, ALDO/SALT, ALDO/SALT+Eplerenone or ALDO/SALT+AHT. ALDO (0.75 µg/h, Steraloids)RT-qPCR and northern-blot(17)
Deng et al, 2016miR-21PTEN/PI3K/Akt, Caspase-3 Bax and Bcl-2miR-21 decreased H2O2-induced apoptosis by decreasing PTEN/PI3K/Akt signaling. miR-21 downregulated the proapoptosis protein Caspase-3 and Bax, and upregulated Bcl-2c-kitD CSCRT-qPCR, western blot and confocal microscopy(18)
Cheng et al, 2016miR-21TGF-β1 and p-ERK/ERKCelastrol attenuated miR-21 upregulation by TGF-β1 and decreased elevated p-ERK/ERK levels in CFs transfected with miR-21Cardiac fibroblastsWestern blot, RT-qPCR and luciferase reporter assay(19)
Xiao et al, 2016miR-21PDCD4miR-21 inhibited apoptosis pathway through downregulating PDCD4, Restored miR-21/PDCD4 pathway could protect myocardial cells against oxidative stress-related apoptosisH9C2 cardiac cellsWestern blot, RT-qPCR and luciferase activity assay(20)
Tao et al, 2016miR-29aVEGF-A and p-ERK1/2miRNA-29a suppressed cardiac fibrosis and fibroblast proliferation via down-regulating p-ERK1/2 and VEGF-A/MAPK signal pathwayCardiac fibroblastsWestern blot and RT-qPCR(21)
Liu et al, 2017miR-29aAPN and collagen I and IIImiR-29a has a negative correlation with ANP in atherosclerosisBlood sampleRT-qPCR and ELISA(22)
Lu et al, 2018miR-29bSPRY1, MAPK, TNF-α, ROS, NADPH oxidase, CCL2 and CCL5miR-29b suppressed the MAPK signaling pathway through inhibiting SPRY1 at the posttranslational level in atherosclerosisHUVECSWestern blot, luciferase reporter assay, statistical analysis, RT-qPCR and ROS determination(23)
Sassi et al, 2018miR-29Wnt signalingmiR-29 promoted cardiac hypertrophy and fibrosis via derepressesing Wnt signalingCardiac fibroblasts, aorta in patients with aortic valve stenosis and aorta in mice induced by TACRT-qPCR, immunohistochemical analyses and secretome analysis(24)
Panizo et al, 2017miR-29bCTGF, COL1A1 and MMP-2miR-29b inhibited CTGF, COL1A1 and MMP-2cardiomyocytes of the left ventricleWestern blot and RT-qPCR(25)
Heid et al, 2017miR-29Col1a1, Col1a2 and col15a1Upregulation of miR-29 decreased col1a1, col1a2 and col15a1Human cardiac fibroblastsLuciferase reporter assay, Masson/immunohistochemical staining, western blot and RNA sequencing(26)
Chen et al, 2018miR-30aCTGF and collagenmiR-30a inhibited CTGF by directly combining with the 3′-UTR of CTGF, thereby reducing collagen and myocardial fibrosis, which improved cardiac functionYoung adult and old Nfu heartsRT-qPCR(27)
Roca-Alonso et al, 2015miR-30GATA-6, β1AR, β2AR and Giα-2miR-30 expression attenuates the contractile response of cardiomyocytes to βAR stimulation (β1AR, β2AR and Giα-2), which reduced cardiomyocyte contractility DOX sustained miR-30 downregulation in cardiomyocytes via improving GATA-6H9c2 cardiac muscle cell lineNanoString technology, luciferase assays, ROS detection and cAMP accumulation(28)
Lai et al, 2016miR-30eACE2, caspase-3 and Beclin-1Silencing miR-30e reverses the heart-protective effect of ACE2 and induces primary cardiomyocyte apoptosis Overexpression of ACE2 attenuates doxorubicin-mediated pathological signaling of primary cardiomyocytesH9C2 cardiomyocytesRT-qPCR and western blot(29)
van Middendorp et al, 2017miR-133aCTGFmiR-133a negatively regulated CTGF expression SRF and NFATc4, as target genes of miR-133a, did not show significant relation with miR-133a in local hypertrophyIsolated cardiomyocytesRT-qPCR and statistical analysis(30)
Li et al, 2010miR-133aNFATc4Silencing of NFATc4 by miR-133a may contribute to miR-133a-mediated anti-hypertrophyNeonatal rat cardiomyocytesRT-qPCR, western blot and immunostaining(31)
Li et al, 2015miR-133aCaspase-8, caspase-9, caspase-3, Bcl-2 and TAGLN2miR-133a suppressed caspase-8, caspase-9, and caspase-3, but improved Bcl-2 and suppressed TAGLN2 expression via binding to 3′-UTR of TAGLN2 mRNAHypoxic H9c2 cellsBioinformatics analysis and dual-luciferase reporter analysis(32)
Rangrez et al, 2017miR-301aCFL2Overexpression of Cfl2 or knockdown of miR-301a resulted in the activation of SRF signaling and overexpression of miR-301a reduced Cfl2 expressionNRVCMRNA isolation, cDNA synthesis, RT-qPCR and microarray analysis(33)
Dong et al, 2016miR-214Ang-II, COL-I and COL-IIImiR-214 may inhibit collagen synthesis in CFBs induced by Ang II and upregulated miR-214 can inhibit COLI and COLIII Ang II negatively correlates with the expression of miR-214CFBsMasson staining, RT-qPCR and western blot(34)
Chaturvedi et al, 2015miR-455MMP-9miR-455 prevented the downstream detrimental effects of MMP9 that lead to fibrosis and myocyte uncouplingCardiosomes (exosomes from cardiomyocytes)Western blot, RT-qPCR and IHC(35)
Liu et al, 2017miR-135aBcl-2miR-135a positively regulated H2O2-induced apoptosis in H9c2 cells via blocking Bcl-2 proteinH9c2 cellsRNA-mediated gene silencing, RNA extraction, RT-qPCR and western blot(36)
Wang et al, 2016miR-142-3pHMGB1 and TGF-β1/Smad3TGF-β1/Smad3 signaling involved in the miR-142-3p/HMGB1-mediated apoptosis and fibrosis of M6200 cells miR-142-3p inhibits H/R-induced apoptosis and fibrosis by the inhibition of HMGB1 expression partlyM6200 cellsBioinformatics analysis and dual-luciferase reporter assay(37)
Yang et al, 2017miR-410HMGB1 and HSPB1miR-410 may inhibit mitophagy and apoptosis following cardiac I/R injury by repressing HSPB1 activity via directly suppressing HMGB1HACMsDual-luciferase assay(38)
Zhang et al, 2018miR-208aCHD9 and Notch/NFBCHD9 is a direct target of miR-208a, which was also related with Notch/NFB signal pathway during I/R injuryH9c2 cellsRT-qPCR, dual-luciferase activity and western blot,(39)
Fan et al, 2018miR-210HGFUpregulation of HGF was observed among the AMI rats after receiving miR-210 agonistsHUVECRT-qPCR, immunohistochemistry, western blot and statistical analysis(40)
Zhang et al, 2018miR-182-5pCIAPIN1miR-182-5p promoted apoptosis in hypoxia-induced cardiomyocytes via negative regulation of CIAPIN1H9c2 and 293T cells/primary rat cardiac muscle cellsBioinformatic analysis and dual-luciferase reporter assay(41)
Liu et al, 2018miR-132TGF-β1 and smad3miRNA-132 decreased the expression of TGF-β1 and smad3 and increased the antioxidant stress and antiapoptotic ability of H9C2 cellsHF patients' blood/H9C2 cellHE/Masson staining, MTT assay, RT-qPCR western blot and statistical analysis(42)
Zhou et al, 2018miR-184HOXA9 and ANP, BNP, PE and UCA1UCA1 promoted cardiac hypertrophy through competitively binding with miR-184 to enhance the expression of HOXA9. The overexpression of miR-184 lessened the enlarged surface area of cardiomyocytes and the elevated expression of fetal genes (ANP and BNP) induced by PECardiomyocyte isolated from neonatal micePlasmid construction and transfection, RT-qPCR, luciferase reporter analysis and western blot(43)
Rubiś et al, 2016miR-99Akt-1 and EGR-1EGR-1 mediated regulation of miR-99 family that serves a key role in determining the fate of cardiac hypertrophy by regulating Akt-1 signalingExtracellular matrix and serumEndomyocardial biopsy and RT-qPCR(44)
Ji et al, 2018miR-327ITGB3miR-327 represses integrin (ITG)B3, contributing to its effect on cardiac fibrosisCardiac fibroblast Immunohistochemistry, western blot and RT-qPCR(45)
Lu et al, 2018miR-672-5pJUNmiR-672-5p had suppressive effects on cardiac hypertrophy through inhibiting the expression of Jun in cardiomyocytesMyocardial cellsRT-qPCR, luciferase assay and western blot(46)
Wang et al, 2018miR-27bALK5 and Smad-2/3 pathwaymiR-27b inhibited AngII-induced Smad-2/3 phosphorylation, miR-27b ameliorates AF through inactivation of Smad-2/3 pathway by inhibiting ALK5, a receptor of TGF-βMyocardial cellsRT-qPCR, luciferase assay and western blot(47)
Yang et al, 2016miR-22AP-1, Bcl-2/Bax, TNF-α and IL-6miR-22 significantly inhibited AP-1 activity, changed Bcl-2/Bax ratio and suppressed TNF-α and IL-6 induced by H/RNeonatal rat ventricular cardiomyocytesRT-qPCR, western blot, ELISA and EMSA(48)
Zhang et al, 2018miR-22Cav3-PKCε pathwaymiR-22 accelerates cardiac fibrosis through the miR-22-Cav3-PKCε pathway and inhibits angiotensin II-mediated excessive collagen deposition through protein kinase C (PKC)ε inactivationCardiac fibroblasts from the neonatal SD ratsRT-qPCR, western blot, Masson trichrome staining, luciferase reporter assay and immunofluorescence staining(49)
Zheng et al, 2018miR-26a-5pULK1, LC3-I and LC3-IImiR-26a-5p can reduce the expression of ULK1 and collagen I, and decrease the activation of LC3-I to LC3-IIPrimary cardiac fibroblastsDual-luciferase reporter assay, western blot and RT-qPCR(50)
Gu et al, 2018miR-147bKLF13miR-147b inhibits cell viability and promotes apoptosis of rat H9c2 cardiomyocytes via downregulating KLF13 expressionH9c2 cellsLuciferase reporter assay and RT-qPCR(51)
Sun et al, 2017miR-145SGK1, PI3K/AKT signaling pathway and HIF-1amiR-145 could be upregulated by HIF-1a in cardiomyocytes under hypoxic conditions, miR-145 overexpression promoted cell viability, inhibited apoptosis and ROS activity and promoted activation of PI3K/AKT signaling pathway via SGK1 upregulationH9c2 cell line and mouse cardiac muscle cell lineRT-qPCR, western blot and ELISA(52)
Chen et al, 2017miR-200cGATA-4 and Bcl-2miR-200c significantly increased GATA-4 expression. Furthermore, downregulation of miR-200c upregulated the expression of the anti-apoptotic gene Bcl-2CardiomyocyteRT-qPCR, luciferase assay and western blot(53)
Meng et al, 2017miR-363Notch1Inhibition of miR-363 protects cardiomyocytes against hypoxia-induced apoptosis through promotion of Notch1 expression and activation of Notch signalingRat myocardium-derived H9C2 and 293T cellsRT-qPCR, MTT and western blot(54)
Li et al, 2015miR-10a, miR-139b and miR-206TNF, IL-1, IL-6, Cx43 and Rho kinaseTNF, IL-1 and IL-6 downregulate the expression of miR-10a, miR-139b, miR-206 and miR-222, and upregulate the expression of Cx43 and Rho kinase in VSMCs. miR-10a, miR-139b, miR-206 and miR-222 could downregulate the expression of Cx43 and Rho kinaseCardiomyocyte from SD rats fed with a high-fat dietStatistical analyses, ELISA and RT-qPCR(55)
Gallego et al, 2016miR-10bAPAF-1miR-10b inhibition in HL-1 cardiomyocytes induced the overexpression of APAF-1CardiomyocytesTaqMan low density array and RT-qPCR(56)
Huang et al, 2016miR-195Bcl-2 and BDNFmiR-195 promotes ischemic apoptosisc by repressing Bcl-2 and inhibits cardiac function of MI rats BDNF abolished the pro-apoptotic role of miR-195, which was reversed by its scavenger TrkB-FNRVMsRNA extraction, RT-qPCR, luciferase activity assay and cell viability assay(57)
Blumensatt et al, 2017miR-208aAT II Locked-nucleic-acid-mediated inhibition of miR-208a function reversed the detrimental effects induced by AT IIPrimary adult rat cardiomyocytes from Lewis ratsStatistical analysis, and histomorphological and immunohistochemical analysis(58)
Marchand et al, 2016miR-322SIRT4, IGF1R and INSRmiR-322 inhibits the insulin pathway/IGF1R and cyclin D, miR-322 downregulates SIRT4, IGF1R and INSR, which thus decreases Akt phosphorylation and insulin actionCardiomyocytes and heart from C57BL/6 mice fed with high-fat diet (10 weeks)RNA isolation, RT-qPCR, western blot and luciferase assay(59)
Zhong et al, 2016miR-19bPTEN, a-SMS and TGFβRIImiR-19b promotes cardiac fibroblast proliferation and migration by downregulating PTEN, which decreased a-SMS expression by targeting TGFβRIIH9C2 cardiomyocytesWestern blot and RT-qPCR(60)
Pan et al, 2015miR-25Oxidative stress pathways/MCUmiR-25 protects cardiomyocytes against oxidative damage by inhibiting the MCUH9C2 cardiomyocytes exposed to doxorubicinFACS, TUNEL assay, immunoblotting, luciferase reporter assay, western blot and RT-qPCR(61)
Das et al, 2017miR-181a/bPTEN, PI3K ROS and mt-COX1miR-181a/b deficiency inhibits PI3K signaling through upregulation of PTEN. miR-181a/b enhanced damage by overproduction of ROS via inhibiting mt-COX1H9c2 cardiomyocytesMeasurement of sarcomere shortening and Ca2+ transients, mitochondrial swelling assay, western blot and statistical analysis(62)
Palomer et al, 2015miR-146Fos-AP1, MMPs and collagenDownregulation of the Fos-AP-1 pathway by miR-146a can inhibit MMP-9 activity and therefore suppresses hypertrophy of cardiomyocytes and fibrosis of the interstitial substanceAC16 cell line (cardiac muscle cells)Immunoblot analysis, statistical analysis, electrophoretic mobility shift assay and RT-qPCR(63)
Khamaneh et al, 2015miR-155NF-κB inflammatory signaling pathwaysActivation of inflammatory signaling pathways/NF-κBCardiomyocytes from diabetes mellitus type 1 model SD rats administrated with streptozotocinRT-qPCR and statistical analysis(64)
Fang et al, 2015miR-3473binflammatory signaling pathways/TRAF3-NF-κBmiR-3473 negatively regulates TRAF3, a well-known negative regulator of the NF-κB pathway, to enhance NF-κB pathwayBacterial infection model with murine macrophagesRT-qPCR and western blot(65)
Kuwabara et al, 2015miR-451LKB1/AMPK pathwayInduces activation of lipotoxicity through suppression of the LKB1/AMPK pathwayNeonatal cardiomyocytes from C57BL/6 mice feed with high fat diet (20 weeks)/heartDual luciferase reporter assay, western blot, transthoracic echocardiography and statistical analysis(66)

[i] PDCD4, programmed cell death 4; TGFβ-2, transforming growth factor β-2; MCP-1, monocyte chemoattractant protein-1; CSCs, cardiac stem cells; CFs, cardiac fibroblasts; APN, adiponectin; HUVECs, human umbilical vein endothelial cells; TAC, transverse aortic constriction; col1a1, collagen 1A1; col1a2, collagen 1A2; col15a1, collagen 15A1; ARVCMs, adult rat ventricular cardiomyocytes; DOX, doxorubicin; β1AR and β2AR, β1-and β2-adrenoceptor; NRVCM, neonatal rat ventricular cardiomyocytes; CFBs, cardiac fibroblasts; H/R, hypoxia/reoxygenation; I/R, ischemia-reperfusion; HMGB1, high-mobility group box 1 protein; HUVECs, human umbilical vein endothelial cells; CIAPIN1, cytokine-induced anti-apoptotic molecule; PE, phenylephrine; HOXA9, homeobox A9; ANP, atrial natriuretic peptide; BNP, brain natriuretic peptide; EGR-1, early growth response protein 1; ITGB3, integrin β3; JUN, Jun proto-oncogene; AP-1, activator protein 1; AF, atrial fibrosis; Bcl-2, B-cell lymphoma-2; Cav3, Caveolin 3; ULK, Unc-51 like autophagy activating kinase; Cx43, connexin 43; APAF-1, apoptotic protease activating factor-1; BDNF, brain derived neurotrophic factor; IGFR1, insulin-like growth factor 1 receptor; PTEN, phosphatase and tensin homolog deleted on chromosome 10; MCU, mitochondrial calcium uptake; MMP-3, matrix metalloproteinase-3; NF-κB, nuclear factor-κB; TRAFs, TNF receptor associated factors; AMPK, adenosine monophosphate-activated protein kinase; TTC, 2,3,5-triphenyltetrazolium chloride; TUNEL, TdT-mediated dUTP nick-end labeling; ELISA, enzyme linked immunosorbent assay; ROS, reactive oxygen species; CFL2, cofilin-2; IHC, immunohistochemistry; HF, heart failure; UCA1, urothelial carcinoma-associated 1; AT, angiotensin; EMSA, electrophoretic mobility shift assay; TNF, tumor necrosis factor; SD, Sprague-Dawley; PKC, protein kinase C; FACS, fluorescence-activated cell sorting; western blot, western blotting analysis; ALDO, aldosterone; SALT, 1.0% NaCl; AHT, triple antihypertensive therapy (240 mg/kg hydralazine + 75 mg/kg hydrochlorothiazide + 1.5 mg/kg reserpine); KLF, Kruppel-like transcription factor; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; SGK, serum and glucocorticoid induced kinase; ChIP, chromatin immunoprecipitation assay; COL-I, type I collagen; COL-III, type III collagen.

Circulating miRs as diagnostic biomarkers

HF is primarily caused by cardiomyopathy, hypertension, diabetes and CHD, among other causes (15). The different etiology is associated with several miRs.

miRs associated with cardiomyopathy

The cardiomyopathies leading to HF predominantly include DCM and ICM (6771). DCM, characterized by left ventricular dilatation, ventricular wall thinning and diffuse myocardial dysfunction, leads to congestive HF (72) and right ventricular dysfunction (73). These pathological changes result in the transition from compensatory hypertrophy to DCM (74). The heart undergoes continuous remodeling of myocardial cells through transduction of intercellular signals and activation of the transcription and transmission pathways (75). Naga Prasad et al (76) performed reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis on a set of samples used for miR microarray analysis, and identified that hsa-mir-378 (P<0.0055), hsa-mir-001 (P<0.0001), hsa-mir-007 (P<0.0009) and hsa-mir-29b (P<0.0087) were notably decreased in DCM compared with control samples; by contrast, hsa-mir-342 (P<0.0004), hsa-mir-214 (P<0.0001), hsa-mir-125b (P<0.0785), hsa-mir-145 (P<0.0091) and hsa-mir-181b (P<0.0047) were significantly increased in DCM compared with non-failing controls, and may be used to indicate the stage of HF development. Enes Coşkun et al (77) investigated 23 pediatric patients (aged 2–192 months) with isolated idiopathic DCM as the experimental group, and 26 age-matched healthy children with innocent murmur as the control group. Patients with fractional shortening of <25% and with a left ventricular end-diastolic diameter >112% of the predicted dimension were considered to have DCM. The results of RT-qPCR demonstrated that the expression levels of miR-454 and miR-518f were significantly higher in DCM patients compared with those in the control group. Furthermore, the expression levels of 10 miRs (miR-618, miR-875-3p, miR-205, miR-194, miR-302a, miR-147, miR-544, has-miR-99b, miR-155 and miR-218) were notably lower in patients with DCM compared with control subjects, suggesting that they may be used as potential diagnostic biomarkers. Interestingly, Miyamoto et al (78) observed that 2 miRs (hsa-miR-636 and hsa-miR-155) were upregulated and 2 miRNAs (hsa-miR-646 and hsa-miR-639) were downregulated in patients with DCM compared with patients with DCM with recovered ventricular function, which indicated that they may serve as diagnostic as well as prognostic biomarkers. However, further research is required to elucidate the specific underlying mechanisms.

Leger et al (79) and Zeng et al (80) measured left ventricular ejection fraction (LVEF) and the 6-min walk test distance (6MWTD) and CBP/p300 interacting transactivators with ED-rich termini 2 (CITED2), hypoxia-inducible factor-1 (HIF-1) in patients with ICM before and after treatment, and identified that LVEF, 6MWTD, CITED2 and HIF-1 levels were significantly lower in the ICM group compared with those in the control group prior to treatment (P<0.01). The N-terminal pro-B-type natriuretic peptide (NT-proBNP), HIF-1 and miR-182 levels in the ICM group were significantly higher compared with those in the control group (P<0.01). Following 4 months of treatment, the levels of 6MWTD, CITED2 and LVEF in the ICM group were significantly increased, whereas the levels of plasma NT-proBNP, HIF-1 and miR-182 were significantly decreased (P<0.01). Furthermore, the plasma miR-182 level was negatively correlated with CITED2, LVEF and 6MWTD (P<0.05) and positively correlated with HIF-1 (P<0.05) in the ICM group. Therefore, miR-182 is correlated with several indicators of HF, and may be considered to reflect the severity of the disease. Olson and Rooij (81) and Fichtlscherer et al (82) observed upregulation of miR-208a and miR-499 and downregulation of the circulating levels of miR-126, miR-17, miR-92a and the inflammation-associated miR-155 in patients with coronary artery disease compared with healthy controls by qPCR. Similarly, the level of miR-145 in smooth muscle was significantly reduced. By contrast, the levels of cardiac muscle-enriched miRs (miR-133a and miR-208a) tended to be higher in patients with coronary artery disease. Li et al (83) demonstrated a decrease of miR-125a, miR-20a and miR-302d levels in ICM using Deep RNA sequencing. Notably, only 55 miRs were indicated to be consistently increased in ICM and non-ischemic cardiomyopathy (NICM), including miR-21-5p, miR-125b-1-3p and miR-106b-5p, among others. However, 38 miRNAs were downregulated in both ICM and NICM (non-ischemic cardiomyopathy), including miR-20a-5p, miR-17-5p and let-7e-5 (83). The findings suggest that miR-182 appears to be a promising new biomarker for the diagnosis of ICM and DCM in clinical research.

miRs associated with hypertension

Hypertension is an independent risk factor for cardiac and cerebrovascular disease (84). It has been reported that at least 50% of patients with long-term hypertension will likely undergo cardiac remodeling, particularly left ventricular remodeling (85). Myocardial cell hypertrophy is among the primary causes underlying the occurrence of HF (86). Notably, it has been demonstrated that miR-208 can induce cardiac hypertrophy and results in the overexpression of β-myosin heavy chain in myocardial fibrosis (87). Several miRs were indicated to be differentially expressed in hypertension, including miR-296-5p, let-7e and human cytomegalovirus (HCMV)-miR-UL112, as encoded by HCMV in previous studies of the hypertension-associated miR spectrum (8890). Interferon regulatory factor 1, which is involved in the regulation of blood pressure by acting on nitric oxide synthase and vascular angiotensin (Ang) receptor, was demonstrated to be a direct target of HCMV-miR-UL112 (91). However, in hypertension, HCMV titers are considered to reflect the expression level of HCMV-miR-UL 112 (91), which is an independent risk factor for hypertension. HCMV has been reported to inhibit vasodilation by impairing nitric oxide synthase function (92) and causing endothelial cell dysfunction (93). However, further research is warranted due to the elusive association between HCMV infection and endothelial dysfunction.

Kontaraki et al (94,95) reported that upregulated miRs included miR-1 and miR-21, whereas downregulated miRs included miR-9, miR-126, miR-133, miR-143 and miR-145 in the hypertension group compared with the healthy control group. In addition, miR-21, miR-143 and miR-145 were negatively correlated and miR-133 was positively correlated with 24-h ambulatory mean blood pressure, mean diastolic blood pressure and mean pulse pressure in the hypertension group. Furthermore, miR-9 and miR-126 were positively correlated with mean pulse pressure, but the association between miR-9 and left ventricular hypertrophy index was positively correlated with the 24-h ambulatory mean blood pressure and mean diastolic blood pressure. Therefore, this miR may reflect the severity of hypertensive HF.

Dickinson et al (96) reported that the circulating levels of miR-423-5p, miR-106b, miR-20b, miR-223, miR-16 and miR-93 were markedly increased in hypertension-induced HF, which was confirmed via RT-qPCR analysis of plasma RNA from hypertensive rats. These results indicate that several miRs can reflect disease progression to a certain extent, and may be used as biomarkers of hypertensive HF. This suggests that miRs should be detected pre- and post-treatment to reduce the effects of medication on the results of the experiment. Hou et al (97) randomly divided 16 spontaneously hypertensive rats (SHR) into the SHR control (distilled water) and intervention SHR (captopril 10 mg/kg/day) groups. An additional 8 Wistar male rats comprised the normal control groups (captopril 10 mg/kg/day or distilled water for 8 weeks). The expression of miR-137 was detected by RT-qPCR and western blot analysis in rat hearts, and miR-137, Ang II, transforming growth factor (TGF)-β1, Smad3, collagen (Col)-I and Col-III were identified to be more highly expressed in the SHR treatment and SHR control groups than the normal control group (P<0.01 and P<0.05, respectively); by contrast, the levels of miR-137, Ang II, TGF-β1, Smad3, Col-I and Col-III were significantly lower in the normal control groups compared with the SHR control group (P<0.01 and P<0.05, respectively). Thus, miR-137 may promote cardiac remodeling in SHR by upregulation of Ang II and the TGF-β1/Smad3 signaling pathway; in addition, captopril intervention can inhibit miR-137 expression. Therefore, miR-137 not only indicates the presence of high blood pressure, it may also reflect its severity.

Li et al (98) reported that insulin-like growth factor (IGF)-1 prevented diabetes-induced cardiomyopathy via marked anti-apoptotic and anti-fibrotic effects, which are mediated by miR-1. These findings provide a new paradigm for the endocrine effects of IGF-1 in the heart, and suggest that cardiac-specific miR-1 may be a useful biomarker and therapeutic target for diabetes-induced cardiomyopathy. Yang et al (99) observed that miR-505 interfered with the migration of cultured endothelial cells through targeting fibroblast growth factor 18, suggesting that miR-505 may be involved in vascular regeneration. In addition, a group of miRs (miR-92a, miR-130a and miR-195) were demonstrated to be abnormally expressed in hypertensive patients with metabolic syndrome. Notably, miR-92a is differentially expressed in the blood of hypertensive and non-hypertensive patients (100) and may promote miR-mediated intercellular communication (101). Kontaraki et al (94,95) confirmed several types of differentially expressed miRs in an animal model: Myocardial hypertrophy was induced by miR-21, miR-208b and miR-499; the anti-myocardial hypertrophy miRs comprised miR-1, miR-26b and miR-133a, of which miR-1, miR-21, miR-208b and miR-499 were upregulated, whereas miR-26b and miR-133a were downregulated in peripheral blood mononuclear cells from patients with hypertension compared with healthy controls. In patients with hypertension, the degree of left ventricular hypertrophy was negatively correlated with the miR-1 and miR-133 indices, whereas the miR-21, miR-26b, miR-208b and miR-499 indices were positively associated with left ventricular hypertrophy.

miRs associated with diabetic HF

Dickstein (102) reported that the occurrence and development of insulin resistance in HF was correlated with overactivation of the renin-angiotensin-aldosterone system (103,104), disturbance of energy metabolism in the myocardium (105), liver pathology, as well as other factors. It was previously demonstrated that the expression of miR-133 and miR-1 increased significantly in myocardial cells following hyperglycemic injury (106). IGF-1 and IGF-1 receptor are the two target genes of miR-1 (107). Previous studies demonstrated an increasing level of miR-133 and decreasing levels of miR-650, miR-222 and miR-338 in hyperglycemic cardiomyocyte injury (108,109). Greco et al (110) collected biopsies from the peri-infarctual area (border zone) and the non-ischemic remote zone from patients with diabetic HF (D-HF), non-diabetic HF (ND-HF) and the control group. miR expression was measured using RT-qPCR in left ventricular biopsies from 10 patients with D-HF and 19 patients with ND-HF affected by non-end-stage ischemic cardiomyopathy. A total of 17 miRs were revealed to be differentially expressed in patients with D-HF and/or ND-HF when compared with control subjects; in particular, miR-34b, miR-34c, miR-210, miR-199b and miR-372 were upregulated, whereas miR-650 and miR-223 were downregulated. Therefore, miRs may not only be obtained from the blood or serum, but also from tissue biopsies, when the content in the body fluids is low. Nandi et al (111) and Deng et al (112) reported that attenuation of miR-133a in diabetic hearts is associated with the induction of autophagy and hypertrophy. In conclusion, attenuation of miR-133a appears to serve a key role in D-HF and contributes to the exacerbation of diabetes-mediated cardiac autophagy and hypertrophy in patients with HF undergoing left ventricular assist device implantation. Chavali et al (113) used multiplex RT-qPCR in insulin 2 mutant Akita mouse hearts (a diabetic mouse model with heart disease) and observed marked downregulation of miR-744, miR-142-3p, miR-384-3p, miR-494, let-7a, miR-450, miR-338, miR-130, miR-142-3p, miR-148, miR-338, miR-345-3p, miR-433, miR-451, miR-455, miR-500, miR-542-3p and miR-872. By contrast, miR-295 was upregulated in Akita mouse hearts. Therefore, miR-295 may be used as a mammalian-specific miR in early embryonic stages. Increased miR-295 expression was associated with pathological changes in Akita mouse hearts. miR-223, as an anti-inflammatory miR, may reflect the progression of diabetic Ins2+/− Akita heart disease or D-HF. In another study, miR-1 and miR-133A were demonstrated to act as regulators of glucose homeostasis in vitro (113). Notably, miR-133a/b can reduce the expression of glucose transporter 4 and inhibit the uptake of glucose by insulin-induced myocardial cells (114). Furthermore, miR-133a/b targets Kruppel-like transcription factor 15, which is directly involved in this process (108). Two other target genes of miR-133a/b are the human ether-a-go-go-related gene and KCNQI, and these two genes are involved in the regulation of cardiac K+ channels and the presence of long QT syndrome in patients with diabetes (115). The decrease of miR-126 in diabetic microvascular tissues may indicate the severity of diabetic vascular complications (116); however, the expression of miR-126 did not decrease, but was rather significantly increased in patients with coronary atherosclerosis (117). In a mouse model of type 1 diabetes mellitus established by streptozotocin (118), 15 miRs were differentially expressed in the myocardium, among which 10 miRs (miR-195, miR-199a-3P, miR-700, miR-142-3p, miR-24, miR-21, miR-22, miR-499-3p, miR-208a and miR-705) were upregulated, whereas 5 miRs (miR-29a, miR-1, miR-373, miR-143 and miR-20a) were notably downregulated. Histological examination revealed hypertrophy of the myocardial cells in type 1 diabetes mellitus group mice compared with the control group, with a disorderly arrangement and enlarged nuclei. Notably, the prediction of associated target genes primarily involves cell growth, differentiation, proliferation, collagen fiber growth, apoptosis and angiogenesis.

miRs of HF in CHD

CHD is a multi-gene genetic disease resulting from structural or functional cardiovascular abnormalities present at birth that are caused by congenital abnormalities (119). Disrupted miR expression may result in CHD via specific protein regulation. miR-133 and miR-1 are present in the same transcription unit (120); miR-1 is the most abundant miR and is highly conserved in human myocardial cells (121). Mature miR-1-1 and miR-1-2 have the same gene sequence; the miR-13 family includes miR-133a-1, miR-133a-2 and miR-133b (122,123). During heart development, the deletion or mutation of the essential gene Hand2 of muscle precursor cells in early embryonic development may lead to cardiac hypoplasia and even cardiac arrest (124). Mukai et al (125) revealed that miR-486-3p, miR-155-5p and miR-486-5p were increased in patients with cyanotic heart disease compared with those without heart disease. Furthermore, let-7e-5p and miR-1260a were decreased in patients with early-stage acyanotic heart disease compared with those without heart disease, suggesting that these miRs may be used for early diagnosis.

Zhao et al (126) reported that the expression of miR-1-2 was upregulated in myocardial and skeletal muscle cells. Overexpression of miR-1 during cardiac development may inhibit ventricular myocyte dilatation. It was also demonstrated that miR-1-2 targets the Hand2 gene, which may block Hand2 protein synthesis and regulate cardiac morphogenesis (127); its abnormal expression may even lead to CHD (127). Another study reported that the mouse phenotypes were almost normal with deletion of either miR-133a-1 or miR-133a-2, but the synchronous lack of these two miRs led to a fatal ventricular septal defect in approximately half of the mice during the embryonic period (128). Thus, miR-133 can promote myoblast proliferation, and miR-1 can stimulate myogenic differentiation. Therefore, miR-1 and miR-133 exhibit a dialectical association, and abnormalities may lead to the development of CHD.

Chen and Li (129) quantified the levels of miR-19a by RT-qPCR in the plasma of 30 patients with CHD, and changes in the levels of miR-19a, miR-130a and miR-27b were also confirmed using RT-qPCR. The levels of miR-19a, miR-198, miR-130a and miR-27b were significantly increased in patients suffering from pulmonary arterial hypertension induced by CHD. These observations suggest that circulating miR-19a may be a novel biomarker for the diagnosis of pulmonary arterial hypertension induced by CHD.

The abovementioned data summarize the differences in expression of miRs in patients with HF (including cardiomyopathy, hypertension, D-HF and CHD). Their clinical significance as HF biomarkers were analyzed (Table II).

Table II.

Expression of miRs in different types of heart failure.

Table II.

Expression of miRs in different types of heart failure.

Author, yearHeart failure typemiR expression levelDetection meansSource(Refs.)
Leger et al, 2013ICMmiR-361 ↑ in ICM groupRT-qPCRSerum(79)
Zeng et al, 2017ICMmiR-182 ↑ in coronary artery disease group compared with the healthy control groupRT-qPCRPlasma/serum(80)
Olson and Rooij, 2014ICMmiR-208 and miR-499 ↑ in ICM groupRT-qPCRSerum(81)
Fichtlscherer et al, 2010ICMmiR-126, miR-17, miR-92 and miR-155 ↓ in ICMRT-qPCRSerum and blood(82)
Li et al, 2018ICMmiR-125a, miR-20a and miR- 302d ↓ only in ICMRT-qPCR and deep RNA sequencingSerum(83)
Li et al, 2018ICMmiR-20a-5p, miR-17-5p and let-7e-5 ↓ in ICM and NICMRT-qPCR and deep RNA sequencingSerum(83)
Li et al, 2018ICMmiR-21-5p, miR-125b-1-3p and miR-106b-5p ↑ in ICM and NICMRT-qPCR and deep RNA sequencingSerum(83)
Naga et al, 2017DCMhsa-miR-214, hsa-miR-342, hsa-miR-125b, hsa-miR-181b and hsa-miR-145 ↑ in the DCM compared with controlsRT-qPCRSerum(76)
Naga Prasad et al, 2017DCMhsa-miR-1, hsa-miR-29b, hsa-miRNA-7, hsa-miR-378 ↓ in the DCM compared with controlsRT-qPCRSerum(76)
Enes Coşkun et al,DCMmiR-454 and miR-518f ↑ in DCMRT-qPCRSerum(77)
Enes Coşkun et al, 2016DCMmiR-618, miR-875-3p, miR-205, miR-194, miR-302a, miR-147, miR-544, has-miR-99b, miR-155 and miR-218 ↓ in DCMRT-qPCRSerum(77)
Miyamoto et al, 2015DCMhsa-miR-636 and hsa-miR-155 ↑ in the DCMRT-qPCRSerum(78)
Miyamoto et al, 2015DCMhsa-miR-639 and hsa-miR-646 ↓ in the DCMRT-qPCRSerum(78)
Ding et al, 2017HHFmiR-296-5p, let-7e and hcmv-miR-UL112 ↑ in the HHF groupRT-qPCRSerum(91)
Kontaraki et al, 2013HHFmiR-1, miR-21, miR-208b and miR-499 ↑ in the HHF groupRT-qPCRSerum(94)
Kontaraki et al, 2013HHFmiR-26b, miR-133a, miR-9, miR-126, miR-133, miR-143 and miR-145↓ in the HHF groupRT-qPCRSerum(95)
Dickinson et al, 2013HHFmiR-16, miR-20b, miR-93, miR-106b, miR-223 and miR-423-5p ↑ in the HHF groupRT-qPCR   Plasma/serum(96)
Hou et al, 2016HHFmiR-137 ↑ in SHR treatment group vs. SHR control group/SHR control group compared with the normal control groupRT-qPCRSerum(97)
Latronico et al, 2007DiabeticmiR-133 ↑ in diabetic heart failure compared with control heart failureRT-qPCRSerum(109)
Latronico et al, 2007Diabetic heart failureLevel of miR-650, miR-222 and miR-338 ↓in diabetic heart failureRT-qPCRSerum(109)
Greco et al, 2012Diabetic heart failuremiR-650 and miR-223 ↓in diabetic heart failureRT-qPCRBody tissue(110)
Greco et al, 2012Diabetic heart failuremiR-34b-34c, miR-210, miR-199b and miR-372 ↑ in D-HF and ND-HF compared with control groupRT-qPCRBody tissue(110)
Nandi et al, 2015Diabetic heart failuremiR-133a ↓ in diabetic heart failure introduced by insulin2 mutant (Ins2+/61) Akita heart diseaseRT-qPCRSerum(111)
Deng et al, 2017Diabetic heart failuremiR-24 ↓ in diabetic heart failureRT-qPCRSerum(112)
Chavali et al, 2014Diabetic heart failuremiR-295 ↑ in AkitaRT-qPCRSerum(113)
Chavali et al, 2014Diabetic heart failuremiR-126, miR-222, miR-130a, miR-142-3p, miR-148, miR-338, miR-345-3p, -miR-384-3p, miR-433, miR-450, miR-451, miR-455, miR-499, miR-500, miR-542-3p, miR-744 and miR-872 ↓ in diabetic heart failureRT-qPCRSerum(113)
van Solingen et al, 2012Diabetic heart failuremiR-126 ↓ in diabetic microvascular tissuesRT-qPCRDiabetic micro-vascular tissues(116)
Fichtlscherer et al, 2010Diabetic heart failuremiR-126 ↑ in patients with coronary atherosclerosisRT-qPCRSerum(117)
Škrha et al, 2015Diabetic heart failure   miR-29a, miR-1, miR-373, miR-143 and miR-20a ↓ in diabetic heart failureRT-qPCRSerum(118)
Škrha et al, 2015Diabetic heart failuremiR-195, miR-199a-3P, miR-700, miR-142-3p, miR-24, miR-21, miR-22, miR-499-3p, miR-208a and miR-705 ↑ in diabetic heart failureRT-qPCRSerum(118)
Li et al, 2018CHDmiR-29 ↑ in patients with CHDRT-qPCRSerum(83)
Mukai et al, 2017CHDmiR-486-3p, miR-155-5p and miR-486-5p ↑ in congenital cyanotic heart diseaseRT-qPCR and microarraysSerum(125)
Mukai et al, 2017CHDmiR-133a-2, let-7e-5p and miR-1260a ↓ in congenital cyanotic heart diseaseRT-qPCR and microarraysSerum(125)
Chen and Li, 2017CHDmiR-19a, miR-198, miR-130a and miR-27b ↑ in patients with CHDRT-qPCRSerum(129)

[i] miR, microRNA; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; HHF, hypertensive heart failure; DCM, dilated cardiomyopathy; CHD, congenital heart disease; ICM, ischemic cardiomyopathy; NICM, nonischemic cardiomyopathy; SHR, spontaneously hypertensive rats; D-HF, diabetic heart failure; ND-HF, nondiabetic heart failure; ↓, downregulation; ↑, upregulation.

Limitations of miRs as biomarkers of HF

Establishing an accurate, reliable circulating miR system for HF diagnosis, prognosis and prediction of response to treatment is challenging, from sample collection and processing to data analysis. First, overlapping between various failure mechanisms leads to difficulties in assessing which mechanisms underlie the expression changes in circulating miRs. Second, serum or plasma are the first choices for sample selection and handling, but the level of circulating biomarker miRs was low, which to some degree impedes the detection of miRs (130). Serum hemolysis may result in waste of samples (131). Furthermore, the serum level of miRs was higher than for circulating plasma, indicating that serum samples can prevent potential interference caused by platelets and leukocytes during sample preparation (132). Therefore, use of the same type of material and synchronous sampling is important for the patient and control groups, as well as a standard scheme to avoid sample hemolysis, minimizing differences between patient selection and classification. Third, some studies have reported fluctuation of miR levels in patients with HF following treatment (133,134). Blood samples were collected at three stages, namely prior to, during and following treatment. A fourth factor was the choice of measurement platform for miR. As indicated in Fig. 2, all research techniques have advantages and disadvantages, but the most commonly used method is RT-qPCR. This method is more sensitive and more cost-effective compared with other methods, but its primary limitation is the inability to detect new miRs. In addition, the standardization of miR expression level may be difficult, as the expression levels of miRs fluctuate with changes in physiological and pathological conditions. Therefore, standard methods are commonly used for the experiments, including the use of equal amounts of starting material (such as serum or plasma), which is more reliable for endogenous miRs for data normalization.

As observed in the present study, the clinical manifestations of HF caused by expression changes of different miRs are similar, and the changes in miR expression caused by different types of HF may also be similar, reflecting the complexity of miR biology.

Conclusion

As described in Fig. 3, the expression of miR-145 was upregulated and the expression of miR-147 and miR-7 was downregulated in DCM, ultimately inhibiting cardiomyocyte apoptosis. The upregulation of miR-181 inhibited oxidative stress. Furthermore, upregulation of miR-214 and downregulation of miR-29b attenuated cardiomyocyte fibrosis, which may be a late regulatory mechanism. By contrast, upregulation of miR-155 promotes cardiomyocyte inflammation, which may be an early regulatory mechanism. The abovementioned miRs appear to be promising potential candidate markers associated with DCM.

In ischemic HF, upregulation of miR-155 intensified cardiomyocyte inflammation, and upregulation of miR-182 promoted apoptosis, which may be an early indicator of this condition. Upregulation of miR-21 alleviated apoptosis via negative feedback regulation. Thus, miR-21 may be a late-age indicator in ischemic HF.

In hypertensive HF, downregulation of miR-133 inhibited cardiomyocyte hypertrophy and promoted cardiomyocyte apoptosis, which may be a late-stage decompensation.

In D-HF, upregulation of miR-22 reduced cardiomyocyte fibrosis, apoptosis and inflammation, and downregulation of miR-455 restrained cell fibrosis, which may be a late indicator of diabetic heart failure, whereas the upregulation of miR-195 and miR-142 aggravated apoptosis and miR-451 downregulation exacerbated cardiomyocyte hypertrophy, which may be an early indicator.

In conclusion, miR-155, miR-22 and miR-133 appear to be promising markers of the development, diagnosis and prognosis of HF. However, further research is required to determine whether there is an efficient miR template for application in clinical oncology practice.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81372150 to BHL and grant nos. nos. 91739301 and 91849102 to MH).

Availability of data and materials

Not applicable.

Authors' contributions

MH and BHL designed and conceived the study. YMH, WWL and JW provided advice and assistance. YMH wrote the manuscript. All the authors have contributed to and approved the final version of the manuscript.

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.

References

1 

Cordes KR and Srivastava D: MicroRNA regulation of cardiovascular development. Circ Res. 104:724–732. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Anderson ME, Brown JH and Bers DM: CaMKII in myocardial hypertrophy and heart failure. J Mol Cell Cardiol. 51:468–473. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Yang J, Savvatis K, Kang JS, Fan P, Zhong H, Schwartz K, Barry V, Mikels-Vigdal A, Karpinski S, Kornyeyev D, et al: Targeting LOXL2 for cardiac interstitial fibrosis and heart failure treatment. Nat Commun. 7:137102016. View Article : Google Scholar : PubMed/NCBI

4 

Kawakami H, Kubota Y, Takeno S, Miyazaki Y, Wada T, Hamada R and Nanashima A: Gastrointestinal: Severe congestive heart failure and acute gastric mucosal necrosis. J Gastroenterol Hepatol. 32:9492017. View Article : Google Scholar : PubMed/NCBI

5 

Petrovic D: Cytopathological basis of heart failure-cardiomyocyte apoptosis, interstitial fibrosis and inflammatory cell response. Folia Biol (Praha). 50:58–62. 2004.PubMed/NCBI

6 

Orsborne C, Chaggar PS, Shaw SM and Williams SG: The renin-angiotensin-aldosterone system in heart failure for the non-specialist: The past, the present and the future. Postgrad Med J. 93:29–37. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Polyakova V, Loeffler I, Hein S, Miyagawa S, Piotrowska I, Dammer S, Risteli J, Schaper J and Kostin S: Fibrosis in endstage human heart failure: Severe changes in collagen metabolism and MMP/TIMP profiles. Int J Cardiol. 151:18–33. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Romaine SP, Tomaszewski M, Condorelli G and Samani NJ: MicroRNAs in cardiovascular disease: An introduction for clinicians. Heart. 101:921–928. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Liu X, Tong Z, Chen K, Hu X, Jin H and Hou M: The role of miRNA-132 against apoptosis and oxidative stress in heart failure. Biomed Res Int. 2018:34527482018.PubMed/NCBI

10 

Gómez AM, Valdivia HH, Cheng H, Lederer MR, Santanaet LF, Cannel MB, McCune SA, Altschuld RA and Lederer WJ: Defective excitation-contraction coupling in experimental cardiac hypertrophy and heart failure. Science. 276:800–806. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Kumar R, Woo MA, Birrer BV, Macey PM, Fonarow GC, Hamilton MA and Harper RM: Mammillary bodies and fornix fibers are injured in heart failure. Neurobiol Dis. 33:236–242. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Neupane B, Zhou Q, Gawaz M and Gramlich M: Personalized medicine in inflammatory cardiomyopathy. Per Med. 15:127–136. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Dludla PV, Dias SC, Obonye N, Johnson R, Louw J and Nkambule BB: A systematic review on the protective effect of N-acetyl cysteine against diabetes-associated cardiovascular complications. Am J Cardiovasc Drugs. 18:283–298. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Güven Bağla A, Içkin Gülen M, Ercan F, Aşgün F, Ercan E and Bakar C: Changes in kidney tissue and effects of erythropoietin after acute heart failure. Biotech Histochem. 93:340–353. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Lindner K, Haier J, Wang Z, Watson DI, Hussey DJ and Hummel R: Circulating microRNAs: Emerging biomarkers for diagnosis and prognosis in patients with gastrointestinal cancers. Clin Sci (Lond). 128:1–15. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Li R, Geng HH, Xiao J, Qin XT, Wang F, Xing JH, Xia YF, Mao Y, Liang JW and Jia 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

17 

Ball JP, Syed M, Marañon RO, Hall ME, Kc R, Reckelhoff JF, Yanes Cardozo LL and Romero DG: Role and regulation of MicroRNAs in aldosterone-mediated cardiac injury and dysfunction in male rats. Endocrinology. 158:1859–1874. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Deng W, Wang Y, Long X, Zhao R, Wang Z, Liu Z, Cao S and Shi B: miR-21 reduces hydrogen peroxide-induced apoptosis in c-kit+ cardiac stem cells in vitro through PTEN/PI3K/Akt signaling. Oxid Med Cell Longev. 2016:53891812016. View Article : Google Scholar : PubMed/NCBI

19 

Cheng M, Wu G, Song Y, Wang L, Tu L, Zhang L and Zhang C: Celastrol-induced suppression of the MiR-21/ERK signalling pathway attenuates cardiac fibrosis and dysfunction. Cell Physiol Biochem. 38:1928–1938. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Xiao J, Pan Y, Li XH, Yang XY, Feng YL, Tan HH, Jiang L, Feng J and Yu XY: Cardiac progenitor cell-derived exosomes prevent cardiomyocytes apoptosis through exosomal miR-21 by targeting PDCD4. Cell Death Dis. 7:e22772016. View Article : Google Scholar : PubMed/NCBI

21 

Tao H, Chen ZW, Yang JJ and Shi KH: MicroRNA-29a suppresses cardiac fibroblasts proliferation via targeting VEGF-A/MAPK signal pathway. Int J Biol Macromol. 88:414–423. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Liu CZ, Zhong Q and Huang YQ: Elevated plasma miR-29a levels are associated with increased carotid intima-media thickness in atherosclerosis patients. Tohoku J Exp Med. 241:183–188. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Lu Z, Wang F, Yu P, Wang X, Wang Y, Tang ST and Zhu HQ: Inhibition of miR-29b suppresses MAPK signaling pathway through targeting SPRY1 in atherosclerosis. Vascul Pharmacol. 102:29–36. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Sassi Y, Avramopoulos P, Ramanujam D, Grüter L, Werfel S, Giosele S, Brunner A, Esfandyari D, Papadopoulou AS, De Strooper B, et al: Cardiac myocyte miR-29 promotes pathological remodeling of the heart by activating Wnt signaling. Nat Commun. 8:16142017. View Article : Google Scholar : PubMed/NCBI

25 

Panizo S, Carrillo-López N, Naves-Díaz M, Solache-Berrocal G, Martínez-Arias L, Rodrigues-Díez RR, Fernández-Vázquez A, Martínez-Salgado C, Ruiz-Ortega M, Dusso A, et al: Regulation of miR-29b and miR-30c by vitamin D receptor activators contributes to attenuate uraemia-induced cardiac fibrosis. Nephrol Dial Transplant. 32:1831–1840. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Heid J, Cencioni C, Ripa R, Baumgart M, Atlante S, Milano G, Scopece A, Kuenne C, Guenther S, Azzimato V, et al: Age-dependent increase of oxidative stress regulates microRNA-29 family preserving cardiac health. Sci Rep. 7:168392017. View Article : Google Scholar : PubMed/NCBI

27 

Chen L, Ji Q, Zhu H, Ren Y, Fan Z and Tian N: miR-30a attenuates cardiac fibrosis in rats with myocardial infarction by inhibiting CTGF. Exp Ther Med. 15:4318–4324. 2018.PubMed/NCBI

28 

Roca-Alonso L, Castellano L, Mills A, Dabrowska AF, Sikkel MB, Pellegrino L, Jacob J, Frampton AE, Krell J, Coombes RC, et al: Myocardial MiR-30 downregulation triggered by doxorubicin drives alterations in β-adrenergic signaling and enhances apoptosis. Cell Death Dis. 6:e17542015. View Article : Google Scholar : PubMed/NCBI

29 

Lai L, Chen J, Wang N, Zhu G, Duan X and Ling F: MiRNA-30e mediated cardioprotection of ACE2 in rats with Doxorubicin-induced heart failure through inhibiting cardiomyocytes autophagy. Life Sci. 169:69–75. 2017. View Article : Google Scholar : PubMed/NCBI

30 

van Middendorp LB, Kuiper M, Munts C, Wouters P, Maessen JG, van Nieuwenhoven FA and Prinzen FW: Local microRNA-133a downregulation is associated with hypertrophy in the dyssynchronous heart. ESC Heart Fail. 4:241–251. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Li Q, Lin X, Yang X and Chang J: NFATc4 is negatively regulated in miR-133a-mediated cardiomyocyte hypertrophic repression. Am J Physiol Heart Circ Physiol. 298:H1340–H1347. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Li AY, Yang Q and Yang K: miR-133a mediates the hypoxia-induced apoptosis by inhibiting TAGLN2 expression in cardiac myocytes. Mol Cell Biochem. 400:173–181. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Rangrez AY, Hoppe P, Kuhn C, Zille E, Frank J, Frey N and Frank D: MicroRNA miR-301a is a novel cardiac regulator of Cofilin-2. PLoS One. 12:e01839012017. View Article : Google Scholar : PubMed/NCBI

34 

Dong H, Dong S, Zhang L, Gao X, Lv G, Chen W and Shao S: MicroRNA-214 exerts a Cardio-protective effect by inhibition of fibrosis. Anat Rec (Hoboken). 299:1348–1357. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Chaturvedi P, Kalani A, Medina I, Familtseva A and Tyagi SC: Cardiosome mediated regulation of MMP9 in diabetic heart: Role of mir29b and mir455 in exercise. J Cell Mol Med. 19:2153–2161. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Liu N, Shi YF, Diao HY, Li YX, Cui Y, Song XJ, Tian X, Li TY and Liu B: MicroRNA-135a regulates apoptosis induced by hydrogen peroxide in rat cardiomyoblast cells. Int J Biol Sci. 13:13–21. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Wang Y, Ouyang M, Wang Q and Jian Z: MicroRNA-142-3p inhibits hypoxia/reoxygenation-induced apoptosis and fibrosis of cardiomyocytes by targeting high mobility group box 1. Int J Mol Med. 38:1377–1386. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Yang F, Li T, Dong Z and Mi R: MicroRNA-410 is involved in mitophagy after cardiac ischemia/reperfusion injury by targeting high-mobility group box 1 protein. J Cell Biochem. 119:2427–2439. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Zhang S, Zhang R, Wu F and LI X: MicroRNA-208a regulates H9c2 cells simulated ischemia-reperfusion myocardial injury via targeting CHD9 through Notch/NF-kappa B signal pathways. Int Heart J. 59:580–588. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Fan ZG, Qu XL, Chu P, Gao YL, Gao XF, Chen SL and Tian NL: MicroRNA-210 promotes angiogenesis in acute myocardial infarction. Mol Med Rep. 17:5658–5665. 2018.PubMed/NCBI

41 

Zhang Y, Fang J and Ma H: Inhibition of miR-182-5p protects cardiomyocytes from hypoxia-induced apoptosis by targeting CIAPIN1. Biochem Cell Biol. 96:646–654. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Liu X, Tong Z, Chen K, Hu X, Jin H and Hou M: The role of miRNA-132 against apoptosis and oxidative stress in heart failure. Biomed Res Int. 2018:34527482018.PubMed/NCBI

43 

Zhou G, Li C, Feng J, Zhang J and Fang Y: lncRNA UCA1 is a novel regulator in cardiomyocyte hypertrophy through targeting the miR-184/HOXA9 axis. Cardiorenal Med. 8:130–139. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Rubiś P, Totoń-Żurańska J, Wiśniowska-Śmiałek S, Holcman K, Kołton-Wróż M, Wołkow P, Wypasek E, Natorska J, Rudnicka-Sosin L, Pawlak A, et al: Relations between circulating microRNAs (miR-21, miR-26, miR-29, miR-30 and miR-133a), extracellular matrix fibrosis and serum markers of fibrosis in dilated cardiomyopathy. Int J Cardiol. 231:201–206. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Ji Y, Qiu M, Shen Y, Gao L, Wang Y, Sun W, Li X, Lu Y and Kong X: MicroRNA-327 regulates cardiac hypertrophy and fibrosis induced by pressure overload. Int J Mol Med. 41:1909–1916. 2018.PubMed/NCBI

46 

Lu Y and Wu F: A new miRNA regulator, miR-672, reduces cardiac hypertrophy by inhibiting JUN expression. Gene. 648:21–30. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Wang Y, Cai H, Li H, Gao Z and Song K: Atrial overexpression of microRNA-27b attenuates angiotensin II-induced atrial fibrosis and fibrillation by targeting ALK5. Hum Cell. 31:251–260. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Yang J, Chen L, Ding J, Zhang J, Fan Z, Yang C, Yu Q and Yang J: Cardioprotective effect of miRNA-22 on hypoxia/reoxygenation induced cardiomyocyte injury in neonatal rats. Gene. 579:17–22. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Zhang L, Yin H, Jiao L, Liu T, Gao Y, Shao Y, Zhang Y, Shan H, Zhang Y and Yang B: Abnormal downregulation of caveolin-3 mediates the pro-fibrotic action of MicroRNA-22 in a model of myocardial infarction. Cell Physiol Biochem. 45:1641–1653. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Zheng L, Lin S and Lv C: MiR-26a-5p regulates cardiac fibroblasts collagen expression by targeting ULK1. Sci Rep. 8:21042018. View Article : Google Scholar : PubMed/NCBI

51 

Gu M, Wang J, Wang Y, Xu Y, Zhang Y, Wu W and Liao S: MiR-147b inhibits cell viability and promotes apoptosis of rat H9c2 cardiomyocytes via down-regulating KLF13 expression. Acta Biochim Biophys Sin (Shanghai). 50:288–297. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Sun N, Meng F, Xue N, Pang G, Wang Q and Ma H: Inducible miR-145 expression by HIF-1a protects cardiomyocytes against apoptosis via regulating SGK1 in simulated myocardial infarction hypoxic microenvironment. Cardiol J. 25:268–278. 2018.PubMed/NCBI

53 

Chen Z, Zhang S, Guo C, Li J and Sang W: Downregulation of miR-200c protects cardiomyocytes from hypoxia-induced apoptosis by targeting GATA-4. Int J Mol Med. 39:1589–1596. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Meng X, Ji Y, Wan Z, Zhao B, Feng C, Zhao J, Li H and Song Y: Inhibition of miR-363 protects cardiomyocytes against hypoxia-induced apoptosis through regulation of Notch signaling. Biomed Pharmacother. 90:509–516. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Li T, Yang GM, Zhu Y, Wu Y, Chen XY, Lan D, Tian K and Liu LM: Diabetes and hyperlipidemia induce dysfunction of VSMCs: Contribution of the metabolic inflammation/miRNA pathway. Am J Physiol Endocrinol Metab. 308:E257–E269. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Gallego I, Beaumont J, López B, Ravassa S, Gómez-Doblas JJ, Moreno MU, Valencia F, de Teresa E, Díez J and González A: Potential role of microRNA-10b down-regulation in cardiomyocyte apoptosis in aortic stenosis patients. Clin Sci (Lond). 130:2139–2149. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Hang P, Sun C, Guo J, Zhao J and Du Z: BDNF-mediates down-regulation of MicroRNA-195 inhibits ischemic cardiac apoptosis in rats. Int J Biol Sci. 12:979–989. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Blumensatt M, Fahlbusch P, Hilgers R, Bekaert M, Herzfeld de Wiza D, Akhyari P, Ruige JB and Ouwens DM: Secretory products from epicardial adipose tissue from patients with type 2 diabetes impair mitochondrial β-oxidation in cardiomyocytes via activation of the cardiac renin-angiotensin system and induction of miR-208a. Basic Res Cardiol. 112:22017. View Article : Google Scholar : PubMed/NCBI

59 

Marchand A, Atassi F, Mougenot N, Clergue M, Codoni V, Berthuin J, Proust C, Trégouët DA, Hulot JS and Lompré AM: miR-322 regulates insulin signaling pathway and protects against metabolic syndrome-induced cardiac dysfunction in mice. Biochim Biophys Acta. 1862:611–621. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Zhong C, Wang K, Liu Y, Lv D, Zheng B, Zhou Q, Sun Q, Chen P, Ding S, Xu Y and Huang H: miR-19b controls cardiac fibroblast proliferation and migration. J Cell Mol Med. 20:1191–1197. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Pan L, Huang BJ, Ma XE, Wang SY, Feng J, Lv F, Liu Y, Liu Y, Li CM, Liang DD, et al: MiR-25 protects cardiomyocytes against oxidative damage by targeting the mitochondrial calcium uniporter. Int J Mol Sci. 16:5420–5433. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Das S, Kohr M, Dunkerly-Eyring B, Lee DI, Bedja D, Kent OA, Leung AK, Henao-Mejia J, Flavell RA and Steenbergen C: Divergent effects of miR-181 family members on myocardial function through protective cytosolic and detrimental mitochondrial microRNA targets. J Am Heart Assoc. 6(pii): e0046942017.PubMed/NCBI

63 

Palomer X, Capdevila-Busquets E, Botteri G, Davidson MM, Rodríguez C, Martínez-González J, Vidal F, Barroso E, Chan TO, Feldman AM, et al: miR-146a targets Fos expression in human cardiac cells. Dis Model Mech. 8:1081–1091. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Khamaneh AM, Alipour MR, Sheikhzadeh Hesari F and Ghadiri Soufi F: A signature of microRNA-155 in the pathogenesis of diabetic complications. J Physiol Biochem. 71:301–309. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Fang Y, Chen H, Hu Y, Li Q, Hu Z, Ma T and Mao X: Burkholderia pseudomallei-derived miR-3473 enhances NF-κB via targeting TRAF3 and is associated with different inflammatory responses compared to Burkholderia thailandensis in murine macrophages. BMC Microbiol. 16:2832016. View Article : Google Scholar : PubMed/NCBI

66 

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

67 

Cohen-Solal A, Beauvais F and Logeart D: Heart failure and diabetes mellitus: Epidemiology and management of an alarming association. J Card Fail. 14:615–625. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Nargesi AA, Esteghamati S, Heidari B, Hafezi-Nejad N, Sheikhbahaei S, Pajouhi A, Nakhjavani M and Esteghamati A: Nonlinear relation between pulse pressure and coronary heart disease in patients with type 2 diabetes or hypertension. J Hypertens. 34:974–980. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Puntmann VO, Carr-White G, Jabbour A, Yu CY, Gebker R, Kelle S, Hinojar R, Doltra A, Varma N, Child N, et al: T1-mapping and outcome in nonischemic cardiomyopathy: All-cause mortality and heart failure. JACC Cardiovasc Imaging. 9:40–50. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Cahill TJ, Ashrafian H and Watkins H: Genetic cardiomyopathies causing heart failure. Circ Res. 113:660–675. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Ortega A, Roselló-Lletí E, Tarazón E, Molina-Navarro MM, Martínez-Dolz L, González-Juanatey JR, Lago F, Montoro-Mateos JD, Salvador A, Rivera M and Portolés M: Endoplasmic reticulum stress induces different molecular structural alterations in human dilated and ischemic cardiomyopathy. PLoS One. 9:e1076352014. View Article : Google Scholar : PubMed/NCBI

72 

Yeung F, Chung E, Guess MG, Bell ML and Leinwand LA: Myh7b/miR-499 gene expression is transcriptionally regulated by MRFs and Eos. Nucleic Acids Res. 40:7303–7318. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Abraityte A, Lunde IG, Askevold ET, Michelsen AE, Christensen G, Aukrust P, Yndestad A, Fiane A, Andreassen A, Aakhus S, et al: Wnt5a is associated with rightventricular dysfunction and adverse outcome in dilated cardiomyopathy. Sci Rep. 7:34902017. View Article : Google Scholar : PubMed/NCBI

74 

Yamamoto S, Yang G, Zablocki D, Liu J, Hong C, Kim SJ, Soler S, Odashima M, Thaisz J, Yehia G, et al: Activation of Mst1 causes dilated cardiomyopathy by stimulating apoptosis without compensatory ventricular myocyte hypertrophy. J Clin Invest. 111:1463–1474. 2003. View Article : Google Scholar : PubMed/NCBI

75 

Zhang Y, Kanter EM and Yamada KA: Remodeling of cardiac fibroblasts following myocardial infarction results in increased gap junction intercellular communication. Cardiovasc Pathol. 19:e233–e240. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Naga Prasad SV, Gupta MK, Duan ZH, Surampudi VS, Liu CG, Kotwal A, Moravec CS, Starling RC, Perez DM, Sen S, et al: A unique microRNA profile in end-stage heart failure indicates alterations in specific cardiovascular signaling networks. PLoS One. 12:e01704562017. View Article : Google Scholar : PubMed/NCBI

77 

Enes C, oşkun M, Kervancıoğlu M, Öztuzcu S, Yılmaz Coşkun F, Ergün S, Başpınar O, Kılınç M, Temel L and Coşkun MY: Plasma microRNA profiling of children with idiopathic dilated cardiomyopathy. Biomarkers. 21:56–61. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Miyamoto SD, Karimpour-Fard A, Peterson V, Auerbach SR, Stenmark KR, Stauffer BL and Sucharov CC: Circulating microRNA as a biomarker for recovery in pediatric dilated cardiomyopathy. J Heart Lung Transplant. 34:724–733. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Leger KJ, Singh S, Canseco D, VonGrote EC, Karim-Ud-Din S, Collins SC, Thibodeau JT, Mishkin JD, Patel PC, Markham DW, et al: Abstract 13120: Identification of novel circulating microRNAs in ischemic cardiomyopathy utilizing whole blood microRNA profiling. Circulation. 128 Suppl 22:A131202013.

80 

Zeng X, Li X and Wen H: Expression of circulating microRNA-182, CITED2 and HIF-1 in ischemic cardiomyopathy and their correlation. J Clin Cardiol. 33:119–122. 2017.(In Chinese).

81 

Olson E and Rooij EV: Dual targeting of miR-208 and miR-499 in the treatment of cardiac disorders. US Patent 14104886. Filed December 12, 2013; issued. June 26–2014.

82 

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

83 

Li X, Liu CY, Li YS, Xu J, Li DG, Li X and Han D: Deep RNA sequencing elucidates microRNA-regulated molecular pathways in ischemic cardiomyopathy and nonischemic cardiomyopathy. Genet Mol Res. 15:gmr74652016.

84 

Phelan D, Watson C, Martos R, Collier P, Patle A, Donnelly S, Ledwidge M, Baugh J and McDonald K: Modest elevation in BNP in asymptomatic hypertensive patients reflects sub-clinical cardiac remodeling, inflammation and extracellular matrix changes. PLoS One. 7:e492592012. View Article : Google Scholar : PubMed/NCBI

85 

Mohammed SF, Hussain S, Mirzoyev SA, Edwards WD, Maleszewski JJ and Redfield MM: Coronary microvascular rarefaction and myocardial fibrosis in heart failure with preserved ejection fraction. Circulation. 131:550–559. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Shyu KG, Wang BW, Cheng WP and Lo HM: MicroRNA-208a increases myocardial endoglin expression and myocardial fibrosis in acute myocardial infarction. Can J Cardiol. 31:679–690. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Cengiz M, Karatas OF, Koparir E, Yavuzer S, Ali C, Yavuzer H, Kirat E, Karter Y and Ozen M: Differential expression of hypertension-associated microRNAs in the plasma of patients with white coat hypertension. Medicine (Baltimore). 94:e6932015. View Article : Google Scholar : PubMed/NCBI

88 

Fu M, Gao Y, Zhou Q, Zhang Q, Peng Y, Tian K, Wang J and Zheng X: Human cytomegalovirus latent infection alters the expression of cellular and viral microRNA. Gene. 536:272–278. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Stern-Ginossar N, Saleh N, Goldberg MD, Prichard M, Wolf DG and Mandelboim O: Analysis of human cytomegalovirus-encoded microRNA activity during infection. J Virol. 83:10684–10693. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Li S, Zhu J, Zhang W, Chen Y, Zhang K, Popescu LM, Ma X, Lau WB, Rong R, Yu X, et al: Signature microRNA expression profile of essential hypertension and its novel link to human cytomegalovirus infection. Circulation. 124:175–184. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Ding M, Wang X, Wang C, Liu X, Zen K, Wang W, Zhang CY and Zhang C: Distinct expression profile of HCMV encoded miRNAs in plasma from oral lichen planus patients. J Transl Med. 15:1332017. View Article : Google Scholar : PubMed/NCBI

92 

Kellawan JM, Johansson RE, Harrell JW, Sebranek JJ, Walker BJ, Eldridge MW and Schrage WG: Exercise vasodilation is greater in women: Contributions of nitric oxide synthase and cyclooxygenase. Eur J Appl Physiol. 115:1735–1746. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Dolcino M, Puccetti A, Barbieri A, Bason C, Tinazzi E, Ottria A, Patuzzo G, Martinelli N and Lunardi C: Infections and autoimmunity: Role of human cytomegalovirus in autoimmune endothelial cell damage. Lupus. 24:419–432. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Kontaraki JE, Marketou ME, Zacharis EA, Parthenakis FI and Vardas PE: MiR-1, miR-9 and miR-126 levels in peripheral blood mononuclear cells of patients with essential hypertension associate with prognostic indices of ambulatory blood pressure monitoring. Eur Heart J. 34 Suppl 1:S51582013. View Article : Google Scholar

95 

Kontaraki JE, Marketou ME, Zacharis EA, Parthenakis FI and Vardas PE: Mir-143/mir-145 levels in peripheral blood mononuclear cells associate with ambulatory blood pressure monitoring parameters in patients with essential hypertension. Eur Heart J. 34 Suppl 1:S56562013. View Article : Google Scholar

96 

Dickinson BA, Semus HM, Montgomery RL, Stack C, Latimer PA, Lewton SM, Lynch JM, Hullinger TG, Seto AG and van Rooij E: Plasma microRNAs serve as biomarkers of therapeutic efficacy and disease progression in hypertension-induced heart failure. Eur J Heart Fail. 15:650–659. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Hou YL, LI SL and Liu LL: Effects of MicroRNA-137 and AngII on cardiac remodeling in spontaneously hypertensive rats. Chin J Comp Med. 7–2016.(In Chinese).

98 

Li JZ, Tang XN, Li TT, Liu LJ, Yu SY, Zhou GY, Shao QR, Sun HP, Wu C and Yang Y: Paeoniflorin inhibits doxorubicin-induced cardiomyocyte apoptosis by downregulating microRNA-1 expression. Exp Ther Med. 11:2407–2412. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Yang Q, Jia C, Wang P, Xiong M, Cui J, Li L, Wang W, Wu Q, Chen Y and Zhang T: MicroRNA-505 identified from patients with essential hypertension impairs endothelial cell migration and tube formation. Int J Cardiol. 177:925–934. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Li Y, Wu H, Zhu M, Shelat H, Qu J, Zheng M, Yuan J, Yuan G, Xu J, Wang H and Geng YJ: Insulin-like growth factor prevents diabetes induced cardiomyopathy mediated by MICRORNA-1. J Am College Cardiol. 55:A21.E1962010. View Article : Google Scholar

101 

Finn NA, Eapen D, Manocha P, Al Kassem H, Lassegue B, Ghasemzadeh N, Quyyumi A and Searles CD: Coronary heart disease alters intercellular communication by modifying microparticle-mediated microRNA transport. FEBS Lett. 587:3456–3463. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Dickstein K: Is substantial renal dysfunction in patients with heart failure no longer a contraindication for RAS inhibition? The power of a large, high-quality registry to illuminate major clinical issues. Eur Heart J. 36:2279–2280. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Shang F, Wang SC, Hsu CY, Miao Y, Martin M, Yin Y, Wu CC, Wang YT, Wu G, Chien S, et al: MicroRNA-92a mediates endothelial dysfunction in CKD. J Am Soc Nephrol. 28:3251–3261. 2017. View Article : Google Scholar : PubMed/NCBI

104 

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

105 

Wang C, Fan F, Cao Q, Shen C, Zhu H, Wang P, Zhao X, Sun X, Dong Z, Ma X, et al: Mitochondrial aldehyde dehydrogenase 2 deficiency aggravates energy metabolism disturbance and diastolic dysfunction in diabetic mice. J Mol Med (Berl). 94:1229–1240. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Wong AK, AlZadjali MA, Choy AM and Lang CC: Insulin resistance: A potential new target for therapy in patients with heart failure. Cardiovasc Ther. 26:203–213. 2008. View Article : Google Scholar : PubMed/NCBI

107 

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

108 

Horie T, Ono K, Nishi H, Iwanaga Y, Nagao K, Kinoshita M, Kuwabara Y, Takanabe R, Hasegawa K, Kita T and Kimura T: MicroRNA-133 regulates the expression of GLUT4 by targeting KLF15 and is involved in metabolic control in cardiac myocytes. Biochem Biophys Res Commun. 389:315–320. 2009. View Article : Google Scholar : PubMed/NCBI

109 

Latronico MV, Catalucci D and Condorelli G: Emerging role of microRNAs in cardiovascular biology. Circ Res. 101:1225–1236. 2007. View Article : Google Scholar : PubMed/NCBI

110 

Greco S, Fasanaro P, Castelvecchio S, D'Alessandra Y, Arcelli D, Di Donato M, Malavazos A, Capogrossi MC, Menicanti L and Martelli F: MicroRNA dysregulation in diabetic ischemic heart failure patients. Diabetes. 61:1633–1641. 2012. View Article : Google Scholar : PubMed/NCBI

111 

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

112 

Deng X, Liu Y, Luo M and Wu J, Ma R, Wan Q and Wu J: Circulating miRNA-24 and its target YKL-40 as potential biomarkers in patients with coronary heart disease and type 2 diabetes mellitus. Oncotarget. 8:63038–63046. 2017.PubMed/NCBI

113 

Chavali V, Tyagi SC and Mishra PK: Differential expression of dicer, miRNAs, and inflammatory markers in diabetic Ins2+/− Akita hearts. Cell Biochem Biophys. 68:25–35. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Izarra A, Moscoso I, Cañón S, Carreiro C, Fondevila D, Martín-Caballero J, Blanca V, Valiente I, Díez-Juan A and Bernad A: miRNA-1 and miRNA-133a are involved in early commitment of pluripotent stem cells and demonstrate antagonistic roles in the regulation of cardiac differentiation. J Tissue Eng Regen Med. 11:787–799. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Liu H, Yang L, Chen KH, Sun HY, Jin MW, Xiao GS, Wang Y and Li GR: SKF-96365 blocks human ether-à-go-go-related gene potassium channels stably expressed in HEK 293 cells. Pharmacol Res. 104:61–69. 2016. View Article : Google Scholar : PubMed/NCBI

116 

van Solingen C, Bijkerk R, de Boer HC, Rabelink TJ and van Zonneveld AJ: The Role of microRNA-126 in vascular homeostasis. Curr Vasc Pharmacol. 13:341–351. 2015. View Article : Google Scholar : PubMed/NCBI

117 

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

118 

Škrha P, Hajer J, Anděl M, Hořínek A and Korabečná M: miRNA as a new marker of diabetes mellitus and pancreatic carcinoma progression. Cas Lek Cesk. 154:122–126. 2015.(In Czech). PubMed/NCBI

119 

Talmud PJ: How to identify gene-environment interactions in a multifactorial disease: CHD as an example. Proc Nutr Soc. 63:5–10. 2004. View Article : Google Scholar : PubMed/NCBI

120 

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

121 

Wang L, Tian D, Hu J, Xing H, Sun M, Wang J, Jian Q and Yang H: MiRNA-145 regulates the development of congenital heart disease through targeting FXN. Pediatr Cardiol. 37:629–636. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Feng Y, Niu LL, Wei W, Zhang WY, Li XY, Cao JH and Zhao SH: A feedback circuit between miR-133 and the ERK1/2 pathway involving an exquisite mechanism for regulating myoblast proliferation and differentiation. Cell Death Dis. 4:e9342013. View Article : Google Scholar : PubMed/NCBI

123 

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

124 

Shan ZX, Lin QX, Deng CY, Zhou ZL, Zhang XC, Fu YH and Yu XY: Plasmid-mediated miRNA-1-2 specifically inhibits Hand2 protein expression in H9C2 cells. Nan Fang Yi Ke Da Xue Xue Bao. 28:1559–1561. 2008.(In Chinese). PubMed/NCBI

125 

Mukai N, Nakayama Y, Murakami S, Tanahashi T, Sessler DI, Ishii S, Ogawa S, Tokuhira N, Mizobe T, Sawa T and Nakajima Y: Potential contribution of erythrocyte microRNA to secondary erythrocytosis and thrombocytopenia in congenital heart disease. Pediatr Res. 83:866–873. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Zhao Y, Samal E and Srivastava D: Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature. 436:214–220. 2005. View Article : Google Scholar : PubMed/NCBI

127 

Lu CX, Gong HR, Liu XY, Wang J, Zhao CM, Huang RT, Xue S and Yang YQ: A novel HAND2 loss-of-function mutation responsible for tetralogy of Fallot. Int J Mol Med. 37:445–451. 2016. View Article : Google Scholar : PubMed/NCBI

128 

Ferreira LR, Frade AF, Santos RH, Teixeira PC, Baron MA, Navarro IC, Benvenuti LA, Fiorelli AI, Bocchi EA, Stolf NA, et al: MicroRNAs miR-1, miR-133a, miR-133b, miR-208a and miR-208b are dysregulated in chronic chagas disease cardiomyopathy. Int J Cardiol. 175:409–417. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Chen W and Li S: Circulating microRNA as a novel biomarker for pulmonary arterial hypertension due to congenital heart disease. Pediatr Cardiol. 38:86–94. 2017. View Article : Google Scholar : PubMed/NCBI

130 

Patrick DM, Montgomery RL, Qi X, Obad S, Kauppinen S, Hill JA, van Rooij E and Olson EN: Stress-dependent cardiac remodeling occurs in the absence of microRNA-21 in mice. J Clin Invest. 120:3912–3916. 2010. View Article : Google Scholar : PubMed/NCBI

131 

Wang Y, Gu J, Roth JA, Hildebrandt MA, Lippman SM, Ye Y, Minna JD and Wu X: Pathway-based serum microRNA profiling and survival in patients with advanced-stage non-small cell lung cancer. Cancer Res. 73:4801–4809. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Hamam R, Hamam D, Alsaleh KA, Kassem M, Zaher W, Alfayez M, Aldahmash A and Alajez NM: Circulating microRNAs in breast cancer: Novel diagnostic and prognostic biomarkers. Cell Death Dis. 8:e30452017. View Article : Google Scholar : PubMed/NCBI

133 

Duttagupta R, Jiang R, Gollub J, Getts RC and Jones KW: Impact of cellular miRNAs on circulating miRNA biomarker signatures. PLos One. 6:e207692011. View Article : Google Scholar : PubMed/NCBI

134 

Sassi Y, Avramopoulos P, Ramanujam D, Grüter L, Werfel S, Giosele S, Brunner AD, Esfandyari D, Papadopoulou AS, De Strooper B, et al: Cardiac myocyte miR-29 promotes pathological remodeling of the heart by activating Wnt signaling. Nat Commun. 8:16142017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang YM, Li WW, Wu J, Han M and Li BH: The diagnostic value of circulating microRNAs in heart failure (Review). Exp Ther Med 17: 1985-2003, 2019
APA
Huang, Y., Li, W., Wu, J., Han, M., & Li, B. (2019). The diagnostic value of circulating microRNAs in heart failure (Review). Experimental and Therapeutic Medicine, 17, 1985-2003. https://doi.org/10.3892/etm.2019.7177
MLA
Huang, Y., Li, W., Wu, J., Han, M., Li, B."The diagnostic value of circulating microRNAs in heart failure (Review)". Experimental and Therapeutic Medicine 17.3 (2019): 1985-2003.
Chicago
Huang, Y., Li, W., Wu, J., Han, M., Li, B."The diagnostic value of circulating microRNAs in heart failure (Review)". Experimental and Therapeutic Medicine 17, no. 3 (2019): 1985-2003. https://doi.org/10.3892/etm.2019.7177