Upregulated microRNA-214 enhances cardiac injury by targeting ITCH during coxsackievirus infection

  • Authors:
    • Zhi‑Gang Chen
    • Hui Liu
    • Jun‑Biao Zhang
    • Shao‑Li Zhang
    • Li‑Hua Zhao
    • Wan‑Qian Liang
  • View Affiliations

  • Published online on: March 24, 2015     https://doi.org/10.3892/mmr.2015.3539
  • Pages: 1258-1264
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Viral myocarditis (VM), a severe clinical condition characterized by cardiac inflammation, is most frequently induced as a result of coxsackievirus infection. Evidence suggests that microRNAs may have significant roles in the progression of cardiac injury during coxsackievirus infection. Concurrently, microRNA (miR)‑214 was found to be upregulated in the plasma and myocardial cells during this process. In the present study, eight candidate miRNAs, the functions of which are associated with myocarditis, were selected and their expression levels were evaluated by reverse transcription-quantitative polymerase chain reaction. miR‑146b and miR‑214 were found to have significantly upregulated expression levels in the heart tissues of patients with VM compared with those of the control subjects. Predictions via the use of online bioinformatics tools and confirmed by dual‑luciferase assay and western blot analysis, revealed that ITCH, an NF‑κB signaling suppressor, was a target gene of miR‑214. To investigate the biological function of miR‑214, tumor necrosis factor‑α and interleukin‑6 expression levels were evaluated in HeLa cell culture supernatant. The results revealed that miR‑214 overexpression enhanced the expression of the two cytokines. In addition, the function of miR‑214 was partially rescued by ITCH overexpression. Subsequently, concurrent results were obtained following experiments in murine cardiac myocytes. In conclusion, the results of the present study demonstrated that miR‑214 contributed to the adverse inflammatory response to viral infection of the heart during coxsackievirus infection and is therefore a potential therapeutic target for the treatment of viral myocarditis.

Introduction

Myocarditis is an inflammation of the heart muscle that is most often due to enteroviral infection. Coxsackievirus B3 (CVB3) is hypothesized to be the most common causative agent underlying myocarditis in humans. Viral myocarditis (VM) affects 5–20% of the human population, and is potentially fatal among infants as well as children (1,2). Although VM is frequently resolved among older individuals, it may develop into chronic myocarditis and/or dilated cardiomyopathy, leading to cardiac failure (35). The pathogenesis of VM is based on adverse immune responses evoked by infection of the cardiac muscle with cardiotropic viruses, which leads to viral elimination as well as cardiac myocyte destruction, reparative fibrosis and heart failure (6). Due to the current dearth of effective therapies for the treatment of myocarditis, further studies of the molecular mechanisms underlying these autodestructive inflammatory signaling pathways within the immune system are required (7).

MicroRNAs (miRNAs) are short, noncoding RNA sequences that function as key gene regulators, modulating gene expression at the post-transcriptional level by targeting the 3′-untranslated region (3′UTR) of messenger RNA sequences (8). Gene expression studies have demonstrated that miRNAs are differentially expressed in heart disease, and loss-of-function studies in mice have confirmed that miRNAs are able to regulate multiple cell processes essential to heart function (9,10). miRNAs have significant functions in the maintenance of normal human physiological conditions, and abnormal miRNA expression has been associated with numerous human diseases, varying from psychiatric disorders to types of malignant cancer (1113).

Recently, a group reported that disturbed miRNA expression, including that of miR-155, -21 and -146b, was associated with VM pathogenesis (10). It has also been hypothesized that miRNAs, for example miR-208b and miR-499-5p, may be used as novel biomarkers for VM diagnosis (14). In the present study, the expression levels of eight candidate miRNAs which may be involved in the pathogenesis of VM were evaluated, in order to determine which were significantly altered. Subsequently, bioinformatics tools were used to predict target genes of the identified dysregulated miRNAs, and the results were verified by dual-luciferase assay and western blot analysis. The biological functions of the identified miRNAs were also studied in mouse cardiac myocytes. NF-κB signaling was upregulated only by overexpressed miR-214, but not miR-146a. Thus, miR-214 is associated with myocardial injury during VM.

Materials and methods

Patients

All human material was obtained from the First Affiliated Hospital of Xinxiang Medical University (Weihui, China). Samples were stored at -80°C immediately following collection and were available for research purposes in accordance with the Declaration of Helsinki. The study was approved by the ethics committee of The First Affiliated Hospital of Xinxiang Medical University (Weihui, China). Informed consent was obtained from the patients or their families. miRNA expression was evaluated in right ventricular septal specimens from patients with myocarditis at the acute stage (n=8), within 3 months after the disease had been diagnosed, as well as a clinical history of myocarditis and confirmed viral presence in cardiac biopsies. The control subjects (n=8) comprised age-matched patients presenting with unexplained ventricular tachy-arrhythmias, but with normal ejection fractions and no identified systemic or cardiac inflammation or viral presence at the time of biopsy.

Cell culture

HeLa cells were cultured in Dulbecco’s modified Eagle medium supplemented with 10% fetal bovine serum (HyClone, Thermo Fisher Scientific, Waltham, MA, USA), 100 IU/ml penicillin and 10 mg/ml streptomycin. Cells were maintained at 37°C in a 5% CO2 atmosphere.

Purification of neonatal murine cardiac myocytes

Six specific pathogen free BALB/c mice (8–10 weeks old) in their first day of pregnancy were were purchased from Shanghai Laboratory Animal Centre (Chinese Academy of Science, Shanghai, China). All of the mice were housed under pathogen free conditions. The mice were maintained in a controlled environment at a temperature of 25±1°C and relative humidity of 50–60%. The environment was artificially illuminated (fluorescent lights), and the mice underwent a 12:12 h light/dark cycle and were fed a standard chow diet. The neonatal mice (postnatal 0–3 days) were sacrificed by cervical dislocation. Cardiac myocytes obtained from neonatal mice were prepared as previously described (15). Briefly, the mouse hearts were removed, minced finely and stepwise enzymatic digestion was performed using 0.25% trypsin-0.02% EDTA. The dissociated cells were subsequently washed with complete basal Eagle’s medium and the endothe-lial cells and fibroblasts were removed by two sequential 1 h adsorptions to plastic flasks at 37°C. Non-adherent myocytes were removed, washed once, resuspended in complete basal medium and plated into tissue culture wells. Following 48 h of incubation, the myocytes had attached to the plastic tissue culture wells, and >95% of the cells were identified as cardiac myocytes according to observations regarding their shape and beating activity. The cultured cells were subsequently evaluated as described below. The experiment was approved by the ethical committee of The First Affiliated Hospital of Xinxiang Medical University.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR analysis was used to examine the relative expression levels of eight candidate miRNAs. Total RNA was extracted from the cell or tissue samples with TRIzol (Invitrogen Life Technologies, Carlsbad, CA, USA), according to the manufacturer’s instructions. Eight candidate miRNAs (miR-214, miR-155, miR-146b, miR-208b, miR-21, miR-499, miR-375 and miR-125b) were selected, which had previously been reported to be associated with VM pathogenesis (10,14,16,17), and their expression levels were evaluated using TaqMan miRNA RT-Real Time PCR (Applied Biosystems, Foster City, CA, USA). Single-stranded complementary DNA was synthesized using the TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems) and subsequently amplified with the TaqMan Universal PCR Master mix (Applied Biosystems), alongside the miRNA-specific TaqMan MGB probes (Applied Biosystems). The reaction mixture was incubated at 95°C for 30 sec, followed by 40 cycles of 95°C for 8 sec and 60°C for 30 sec. The U6 small nuclear RNA was used for normalization. Each sample, within each group, was evaluated in triplicate and each experiment was repeated ≥3 times.

Target gene prediction

Potential target genes were searched for using the online bioinformatics tool TargetScan Release 6.2 (http://www.targetscan.org/) (18). The results suggested that ITCH mRNA may be directly targeted by miR-214.

Dual luciferase assay

The full length of ITCH 3′UTR was cloned into a pGL3 vector (Promega Corp., Madison, WI, USA), downstream of the firefly luciferase coding region in order to produce the luciferase reporter vector. For the luciferase reporter assays, HeLa cells were seeded into 48-well plates. miRNA mimics, miRNA antagonists or their corresponding controls and luciferase reporter vectors were co-transfected using Lipofectamine® 2000 (Invitrogen Life Technologies). The pRT-TK vector, which expresses Renilla luciferase was used as a transfection control. Cells were harvested following two days of cotransfection and evaluated with the Dual-Luciferase assay (Promega Corp.). Each transfection was performed in triplicate in three independent experiments. The results are expressed as relative luciferase activity: (Firefly luciferase/Renilla luciferase). A reporter vector containing mutant ITCH 3′UTR was constructed using Fast Mutagenesis system (Beijing TransGen Biotech Co., Ltd, Beijing, China), to identify the binding region of miR-214. The binding region sequence of miR-214 (CCUGCUG) was replaced with CAUACUA. The wild type or mutant ITCH 3′UTR reporter vector was then transfected into HEK293T cells (China Infrastruture of Cell Line Resources, Beijing, China) alongside the miR-214 mimic or inhibitor using lipofectamine 2000. A total of 48 h post-transfection, the cells were lysed by incubation in Passive Lysis buffer on ice for 15 min and luciferase activity was detected using the Dual-Luciferase Assay kit.

Western blot analysis

Protein extracts were boiled in SDS/β-mercaptoethanol sample buffer, and 20 μg samples were loaded onto 8% polyacrylamide gels. The proteins were separated by electrophoresis, and blotted onto poly-vinylidene difluoride membranes (Amersham Pharmacia Biotech, St. Albans, Herts, UK) via electrophoretic transfer. The membrane was incubated with rabbit polyclonal anti-human ITCH antibody (1:500; cat. no. ab31097; Abcam, Cambridge, MA, USA) and mouse monoclonal anti-human β-actin antibody (1:1,000; cat. no. sc-47778; Santa Cruz Biotechnology Inc., Dallas, TX, USA) for 1 h at 37°C. The membrane was subsequently incubated with horseradish peroxidase-conjugated goat anti-rabbit (1:5,000; cat. no. sc-2004) or goat anti-mouse (1:10,000; cat. no. sc-2060) secondary antibodies (Santa Cruz Biotechnology, Inc.) for 1 h at 37°C. Detection was performed via enhanced chemiluminescence using an ECL kit (Pierce Biotechnology Inc., Thermo Fisher Scientific, Rockford, IL, USA). β-actin was used as a loading control.

Homology analysis

The sequences of human and mouse ITCH gene were downloaded from GenBank (http://www.ncbi.nlm.nih.gov/genbank/) and analyzed using CLUSTAL X, version 1.83 (http://www.clustal.org/) (19).

Cytokine assays

The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and monocyte chemoattractant protein (MCP)-1 of cell culture supernatants were determined with ELISA (eBioscience Inc., San Diego, CA, USA) according to the manufacturer’s instructions.

Statistical analysis

Data were analyzed with SPSS Statistical Package version 16 (SPSS, Inc., Chicago, IL, USA). Independent two group’s analyses are used t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-214 and miR-146b are downregulated in the heart tissue of patients with VM

In the present study, the expression levels of eight miRNAs, which were previously reported to exhibit altered expression levels during CVB3 infection, were evaluated in heart tissue samples from patients with CVB3-infected VM. miR-214 and miR-146b were revealed to be significantly upregulated (Fig. 1A and B). Using bioinformatics tools, ITCH, an E3 ubiquitin ligase which functions as an NF-κB suppressor, was identified as a predicted target gene of miR-214 and miR-146b (Fig. 1C).

The expression of ITCH is suppressed by miR-214, but not miR-146b, in HeLa cells

To confirm whether the expression of ITCH was supressed by these miRNAs, the full length (3.6 kb) ITCH 3′UTR was cloned into a pGL3 vector downstream of the luciferase coding region (Fig. 2A). The results of a dual-luciferase assay in HeLa cells indicated that miR-214 significantly supressed firefly luciferase expression by binding to the 3′UTR of ITCH; however, miR-146b did not significantly influence luciferase expression levels (Fig. 2B). To further confirm whether miR-214 repressed ITCH expression, miR-214 was overexpressed and knocked down in HeLa cells, in which ITCH expression was detectable (Fig. 2C). Protein expression levels of ITCH were evaluated by western blot analysis (Fig. 2C), and expression levels of miR-214 and miR-146b were detected by RT-qPCR (Fig. 2D). As shown in Fig. 2B and C, ITCH expression levels were significantly reduced by the miR-214 mimic and slightly upregulated by the miR-214 inhibitor, which indicated that ITCH was a target gene of miR-214.

miR-214 upregulates TNF-α and IL-6 expression by targeting ITCH

The NF-κB family of transcription factors is crucial in the mediation of inflammatory responses. ITCH is a key component of the ubiquitin-editing complex required for termination of the pro-inflammatory activation of the c-Jun N-terminal kinases and NF-κB. Following transfection with miR-214 mimic and CVB3 infection, ITCH expression levels were markedly downregulated, compared with those of cells transfected with the miRNA control (Fig. 3A). To further investigate the biological functions of miR-214 overexpression, the expression levels of TNF-α and IL-6 in cell culture supernatants were examined. As indicated in Fig. 3B, CVB3 infection induced upregulation of TNF-α and IL-6 levels, and following transfection with miR-214 mimic, the concentration of these cytokines was significantly enhanced (P<0.05). Following cotransfection with ITCH expression vector, the expression levels of TNF-α and IL-6 were significantly reduced (Fig. 3C), indicating that miR-214 promoted cell inflammatory responses partially via the suppression of ITCH expression.

miR-214 upregulates TNF-α, IL-1β, IL-6 and MCP-1 expression by targeting ITCH in murine cardiac myocytes

To further investigate this overexpression function in cardiac cells, cardiac myocytes were purified from neonatal mice. Prior to the functional study, the miR-214 binding site in ITCH 3′UTR was identified and the sequences of the human and mouse ITCH 3′UTR were aligned. As exhibited in Fig. 4A, when three nucleotides were mutated, luciferase activity was no longer repressed by miR-214, indicating that the miR-214 binding site was CCUGCUG. Furthermore, this region is highly conserved between human and mouse. Following infection with CVB3, the expression of ITCH was upregulated in a time-dependent manner in those cells transfected with the miRNA control. An analogous effect on ITCH expression was observed in those cells transfected with miR-214 mimic; however, the ITCH protein levels were markedly lower in mouse cardiac myocytes (Fig. 4B). Furthermore, the concentrations of TNF-α, IL-6, IL-1β and MCP-1 in cell culture supernatants were upregu-lated by miR-214 mimic transfection and downregulated by miR-214 antagonist transfection (Fig. 4C).

Discussion

Myocarditis most frequently occurs as a result of infection by enteroviruses, and CVB3 is hypothesized to be the most common causative factor in human myocarditis (6). Despite decades of extensive research, the pathogenesis of VM has remained elusive, and there is currently no effective therapy available for the treatment of this disease (20). Experimental studies have revealed that although CVB3 is able to directly destroy the myocardium (21,22), it is the associated aberrant inflammatory response that is primarily responsible for the induction of myocyte damage (23,24). Additionally, clinical studies have identified enhanced levels of circulating TNF-α, IL-1β, IL-6 and other pro-inflammatory cytokines in patients with myocarditis (25,26). Furthermore, specific immunosup-pressive agents may be used to control the inflammatory response in clinical therapies (27). Therefore, modulation of the inflammatory response represents a potential therapeutic strategy for the treatment of viral myocarditis.

The expression of miRNAs, which function as key gene regulators, was found to be disturbed during CVB3 infection, and certain miRNAs were revealed to have promotional and/or inhibitory roles in the mediation of CVB3 infection. In the present study, eight candidate miRNAs, the functions of which are associated with myocarditis, were selected (10,14,28,29), and their expression was evaluated by RT-qPCR. The results revealed that miR-146b and miR-214 expression levels were significantly upregulated in VM tissues compared with those of the control tissues. Online bioinformatics tools were used to predict ITCH, an NF-κB signaling suppressor, as a target gene of miR-214. This prediction was verified by dual-luciferase assay and western blot analysis. To investigate the biological function of miR-214, TNF-α and IL-6 expression levels were evaluated in HeLa cell culture supernatants. It was demonstrated that the expression levels of TNF-α and IL-6 were upregulated by miR-214 mimic transfection. There was no significant difference in TNF-α or IL-6 expression between the miR-214 knockdown group and that of the control group, although this may have been a result of the low expression levels of miR-214 in HeLa cells. This hypothesis was indirectly confirmed by the experiments performed in murine cardiac myocytes, where the expression of cytokines was significantly reduced by transfection with the miR-214 antagonist.

In conclusion, to the best of our knowledge, the present study was the first to report that the upregulation of miR-214 expression was associated with VM pathogenesis by targeting ITCH. Overexpression of miR-214 upregulated the expression of cytokines and chemokines that enhance myocardial inflammation by weakening NF-κB pathway feedback signaling.

References

1 

Duncan BW, Bohn DJ, Atz AM, French JW, Laussen PC and Wessel DL: Mechanical circulatory support for the treatment of children with acute fulminant myocarditis. J Thorac Cardiovasc Surg. 122:440–448. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Woodruff JF: Viral myocarditis. A review. Am J Pathol. 101:425–484. 1980.PubMed/NCBI

3 

Bowles NE, Richardson PJ, Olsen EG and Archard LC: Detection of Coxsackie-B-virus-specific RNA sequences in myocardial biopsy samples from patients with myocarditis and dilated cardiomyopathy. Lancet. 1:1120–1123. 1986. View Article : Google Scholar : PubMed/NCBI

4 

Kandolf R, Klingel K, Mertsching H, et al: Molecular studies on enteroviral heart disease: patterns of acute and persistent infections. Eur Heart J. 12(Suppl D): 49–55. 1991. View Article : Google Scholar : PubMed/NCBI

5 

Morimoto S, Hiramitsu S, Yamada K, et al: Clinical and pathologic features of chronic myocarditis: four autopsy cases presenting as dilated cardiomyopathy in life. Am J Cardiovasc Pathol. 4:181–191. 1992.PubMed/NCBI

6 

Maier R, Krebs P and Ludewig B: Immunopathological basis of virus-induced myocarditis. Clin Dev Immunol. 11:1–5. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Shauer A, Gotsman I, Keren A, et al: Acute viral myocarditis: current concepts in diagnosis and treatment. Isr Med Assoc J. 15:180–185. 2013.PubMed/NCBI

8 

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

9 

De Rosa S, Curcio A and Indolfi C: Emerging role of microRNAs in cardiovascular diseases. Circ J. 78:567–575. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Corsten MF, Papageorgiou A, Verhesen W, et al: MicroRNA profiling identifies microRNA-155 as an adverse mediator of cardiac injury and dysfunction during acute viral myocarditis. Circ Res. 111:415–425. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Maes OC, Chertkow HM, Wang E and Schipper HM: MicroRNA: implications for alzheimer disease and other human CNS disorders. Curr Genomics. 10:154–168. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Xu J, Li Y, Wang F, et al: Suppressed miR-424 expression via upregulation of target gene Chk1 contributes to the progression of cervical cancer. Oncogene. 32:976–987. 2013. View Article : Google Scholar

13 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in human cancer. Adv Exp Med Biol. 774:1–20. 2013.PubMed/NCBI

14 

Corsten MF, Dennert R, Jochems S, et al: Circulating MicroRNA-208b and MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ Cardiovasc Genet. 3:499–506. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Shen Y, Xu W, Chu YW, Wang Y, Liu QS and Xiong SD: Coxsackievirus group B type 3 infection upregulates expression of monocyte chemoattractant protein 1 in cardiac myocytes, which leads to enhanced migration of mononuclear cells in viral myocarditis. J Virol. 78:12548–12556. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Liu YL, Wu W, Xue Y, et al: MicroRNA-21 and -146b are involved in the pathogenesis of murine viral myocarditis by regulating TH-17 differentiation. Arch Virol. 158:1953–1963. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Tijsen AJ, Pinto YM and Creemers EE: Circulating microRNAs as diagnostic biomarkers for cardiovascular diseases. Am J Physiol Heart Circ Physiol. 303:H1085–H1095. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F and Higgins DG: The CLUSTAL_X windows interface: Flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res. 25:4876–4882. 1997. View Article : Google Scholar

20 

Esfandiarei M and McManus BM: Molecular biology and pathogenesis of viral myocarditis. Annu Rev Pathol. 3:127–155. 2008. View Article : Google Scholar

21 

Klingel K and Kandolf R: The role of enterovirus replication in the development of acute and chronic heart muscle disease in different immunocompetent mouse strains. Scand J Infect Dis Suppl. 88:79–85. 1993.PubMed/NCBI

22 

Fuse K, Chan G, Liu Y, et al: Myeloid differentiation factor-88 plays a crucial role in the pathogenesis of Coxsackievirus B3-induced myocarditis and influences type I interferon production. Circulation. 112:2276–2285. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Leipner C, Grün K, Borchers M and Stelzner A: The outcome of coxsackievirus B3-(CVB3-) induced myocarditis is influenced by the cellular immune status. Herz. 25:245–248. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Calabrese F and Thiene G: Myocarditis and inflammatory cardiomyopathy: microbiological and molecular biological aspects. Cardiovasc Res. 60:11–25. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Matsumori A, Yamada T, Suzuki H, Matoba Y and Sasayama S: Increased circulating cytokines in patients with myocarditis and cardiomyopathy. Br Heart J. 72:561–566. 1994. View Article : Google Scholar : PubMed/NCBI

26 

Levine B, Kalman J, Mayer L, Fillit HM and Packer M: Elevated circulating levels of tumor necrosis factor in severe chronic heart failure. N Engl J Med. 323:236–241. 1990. View Article : Google Scholar : PubMed/NCBI

27 

Schultz JC, Hilliard AA, Cooper LT Jr and Rihal CS: Diagnosis and treatment of viral myocarditis. Mayo Clin Proc. 84:1001–1009. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Xu HF, Ding YJ, Shen YW, et al: MicroRNA-1 represses Cx43 expression in viral myocarditis. Mol Cell Biochem. 362:141–148. 2012. View Article : Google Scholar

29 

Hemida MG, Ye X, Zhang HM, et al: MicroRNA-203 enhances coxsackievirus B3 replication through targeting zinc finger protein-148. Cell Mol Life Sci. 70:277–291. 2013. View Article : Google Scholar

Related Articles

Journal Cover

July-2015
Volume 12 Issue 1

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen ZG, Liu H, Zhang JB, Zhang SL, Zhao LH and Liang WQ: Upregulated microRNA-214 enhances cardiac injury by targeting ITCH during coxsackievirus infection. Mol Med Rep 12: 1258-1264, 2015
APA
Chen, Z., Liu, H., Zhang, J., Zhang, S., Zhao, L., & Liang, W. (2015). Upregulated microRNA-214 enhances cardiac injury by targeting ITCH during coxsackievirus infection. Molecular Medicine Reports, 12, 1258-1264. https://doi.org/10.3892/mmr.2015.3539
MLA
Chen, Z., Liu, H., Zhang, J., Zhang, S., Zhao, L., Liang, W."Upregulated microRNA-214 enhances cardiac injury by targeting ITCH during coxsackievirus infection". Molecular Medicine Reports 12.1 (2015): 1258-1264.
Chicago
Chen, Z., Liu, H., Zhang, J., Zhang, S., Zhao, L., Liang, W."Upregulated microRNA-214 enhances cardiac injury by targeting ITCH during coxsackievirus infection". Molecular Medicine Reports 12, no. 1 (2015): 1258-1264. https://doi.org/10.3892/mmr.2015.3539