Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Can J Cardiol ; 35(4): 480-489, 2019 04.
Article in English | MEDLINE | ID: mdl-30935639

ABSTRACT

BACKGROUND: Long noncoding RNAs (lncRNAs) are emerging as important mediators of cardiac pathophysiology. The aim of the present study is to investigate the effects of lncR-30245, an lncRNA, on cardiac fibrogenesis and the underlying mechanism. METHODS: Myocardial infarction (MI) and transforming growth factor (TGF)-ß1 were used to induce fibrotic phenotypes. Cardiac fibrosis was detected by Masson's trichrome staining. Cardiac function was evaluated by echocardiography. Western blot, quantitative reverse transcription-polymerase chain reaction, and pharmacological approaches were used to investigate the role of lncR-30245 in cardiac fibrogenesis. RESULTS: Expression of lncR-30245 was significantly increased in MI hearts and TGF-ß1-treated cardiac fibroblasts (CFs). LncR-30245 was mainly located in the cytoplasm. Overexpression of lncR-30245 promoted collagen production and CF proliferation. Knockdown of lncR-30245 significantly inhibited TGF-ß1-induced collagen production and CF proliferation. LncR-30245 overexpression inhibited the antifibrotic role of peroxisome proliferator-activated receptor (PPAR)-γ and increased connective tissue growth factor (CTGF) expression, whereas lncR-30245 knockdown exerted the opposite effects. Rosiglitazone, a PPAR-γ agonist, significantly inhibited lncR-30245-induced CTGF upregulation and collagen production in CFs. In contrast, T0070907, a PPAR-γ antagonist, attenuated the inhibitory effects of lncR-30245 small interfering RNA (siRNA) on TGF-ß1-induced CTGF expression and collagen production. LncR-30245 knockdown significantly enhanced ejection fraction and fractional shortening and attenuated cardiac fibrosis in MI mice. CONCLUSION: Our study indicates that the lncR-30245/PPAR-γ/CTGF pathway mediates MI-induced cardiac fibrosis and might be a therapeutic target for various cardiac diseases associated with fibrosis.


Subject(s)
Connective Tissue Growth Factor/metabolism , Heart Diseases/metabolism , PPAR gamma/metabolism , RNA, Long Noncoding , Animals , Benzamides/pharmacology , Cell Proliferation , Collagen/drug effects , Collagen/metabolism , Fibroblasts/metabolism , Fibrosis , Mice, Inbred C57BL , Models, Animal , Myocardial Infarction/metabolism , Pyridines/pharmacology , Rosiglitazone/pharmacology , Stroke Volume , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects
2.
Int J Biol Sci ; 13(1): 22-31, 2017.
Article in English | MEDLINE | ID: mdl-28123343

ABSTRACT

Background: MicroRNAs (miRNAs) have been emerged as important regulator in a multiple of cardiovascular disease, including arrhythmia, cardiac hypertrophy and fibrosis, and myocardial infarction. The aim of this study was to investigate whether miRNA let-7a has antihypertrophic effects in angiotensin II (AngII)-induced cardiac hypertrophy. Methods: Neonatal rat ventricular myocytes (NRVMs) were exposed to AngII for 36 h as a cellular model of hypertrophy; subcutaneous injection of AngII for 2 weeks was used to establish a mouse model of cardiac hypertrophy in vivo study. Cell surface area (CSA) was measured by immunofluorescence cytochemistry; expression of hypertrophy-related genes ANP, BNP, ß-MHC was detected by Real-time PCR; luciferase activity assay was performed to confirm the miRNA's binding site in the calmodulin (CaM) gene; CaM protein was detected by Western blot; the hypertrophy parameters were measured by echocardiographic assessment. Results: The expression of let-7a was decreased in AngII-induced cardiac hypertrophy in vitro and in vivo. Overexpression of let-7a attenuated AngII-induced increase of cell surface area and repressed the increased mRNA levels of ANP, BNP and ß-MHC. Dual-luciferase reporter assay showed that let-7a could bind to the 3'UTR of CaM 1 gene. Let-7a downregulated the expression of CaM protein. In vivo, let-7a produced inhibitory effects on cardiac hypertrophy, including the downregulation of cross-sectional area of cardiomyocytes in mouse heart, the reduction of IVSD and LVPWD, the suppression of hypertrophy marker genes ANP, BNP, ß-MHC mRNA level, and the downregulation of CaM protein level. Conclusions: let-7a possesses a prominent anti-hypertrophic property by targeting CaM genes. The findings provide new insight into molecular mechanism of cardiac hypertrophy.


Subject(s)
Calmodulin/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , 3' Untranslated Regions/genetics , Angiotensin II/pharmacology , Animals , Atrial Natriuretic Factor/genetics , Calmodulin/genetics , Cardiomegaly/chemically induced , Cells, Cultured , MicroRNAs/genetics , Natriuretic Peptide, Brain/genetics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction
3.
Oncotarget ; 8(5): 8752-8764, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28060734

ABSTRACT

BACKGROUND: The aim of the present study was to investigate the role of microRNA (miRNA) let-7a in down-regulation of ß1-adrenoceptors (ß1-AR) and elucidate the underlying mechanism of chronic ischemia heart failure (CIHF) in rats. METHODS AND RESULTS: CIHF model was established by occlusion of coronary artery for 4 weeks. ß1-AR level was obviously down-regulated and let-7a up-regulated in the failing heart 4 weeks after myocardial infarction. Overexpression of let-7a inhibited ß1-AR expression in neonatal rat ventricular cells (NRVCs), which was abolished by anti-let-7a antisense inhibitor. The lentivirus vector containing precursor let-7a (len-pre-let-7a) further down-regulated the reduced ß1-AR level by CIHF and the effect was reversed by len-AMO-let-7a. Len-negative control did not produce any significant influence on ß1-AR expression. Importantly, there exists a negative feedback loop associated with ß1-AR regulation through ß1-AR/cAMP/PKA/GATA4/let-7a/ß1-AR signaling pathway in CIHF. As demonstrated, GATA4 was activated by ß1-AR up-regulation through cAMP-PKA signaling pathway in early phase of ischemia, then GATA4 positively regulated let-7a expression which in turn suppressed ß1-AR expression. CONCLUSIONS: Let-7a regulates ß1-AR expression and forms a negative feedback loop with ß1-AR signaling pathway in ischemic heart failure. This study provides a new insight into the differential expression of ß1-AR in early and later phase of myocardial ischemia.


Subject(s)
Heart Failure/metabolism , MicroRNAs/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction , 3' Untranslated Regions , Animals , Binding Sites , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Feedback, Physiological , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Male , MicroRNAs/genetics , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/pathology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...