Cargando…

Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway

BACKGROUND: MicroRNAs (miRNAs) have been shown to commonly contribute to cardiac hypertrophy (CH). The aim of this study was to test the hypothesis that miR‐200c plays an important role in the progression of CH by targeting myosin light chain kinase (MLCK/MYLK). METHODS AND RESULTS: Cardiac hypertro...

Descripción completa

Detalles Bibliográficos
Autores principales: Hu, Shan, Cheng, Mian, Guo, Xin, Wang, Shun, Liu, Beilei, Jiang, Hong, Huang, Congxin, Wu, Gang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6433679/
https://www.ncbi.nlm.nih.gov/pubmed/30680929
http://dx.doi.org/10.1111/jcmm.14135
_version_ 1783406319000092672
author Hu, Shan
Cheng, Mian
Guo, Xin
Wang, Shun
Liu, Beilei
Jiang, Hong
Huang, Congxin
Wu, Gang
author_facet Hu, Shan
Cheng, Mian
Guo, Xin
Wang, Shun
Liu, Beilei
Jiang, Hong
Huang, Congxin
Wu, Gang
author_sort Hu, Shan
collection PubMed
description BACKGROUND: MicroRNAs (miRNAs) have been shown to commonly contribute to cardiac hypertrophy (CH). The aim of this study was to test the hypothesis that miR‐200c plays an important role in the progression of CH by targeting myosin light chain kinase (MLCK/MYLK). METHODS AND RESULTS: Cardiac hypertrophy was induced by aortic banding (AB) in rats. Cellular hypertrophy in neonatal rat cardiomyocytes (NCMs) was induced by AngII treatment. Echocardiography, histology and molecular measurements were used to assess the results of the experiments. The levels of apoptosis and reactive oxygen species (ROS) were also measured. Quantitative real‐time PCR (qRT‐PCR) and Western blotting were used to measure mRNA and protein levels respectively. The present results showed that miR‐200c expression was increased in response to CH both in vivo and in vitro. The down‐regulation of miRNA‐200c by a specific inhibitor markedly ameliorated CH resulting from AngII treatment, and the mRNA levels of atrial natriuretic peptide, brain natriuretic peptide and β‐myosin heavy chain were simultaneously decreased. Notably, minimal apoptosis and ROS accumulation were identified in AngII‐induced hypertrophic cardiomyocytes. Conversely, the up‐regulation of miR‐200c using specific mimics reversed these effects. Mechanistic investigations demonstrated that the MLCK gene is a direct target of miR‐200c; an increase in miR‐200c levels led to a decrease in the expression of MLCK and its downstream effector, p‐MLC2, while miR‐200c inhibition increased the expression of these proteins. Furthermore, inhibiting MLCK impaired the anti‐hypertrophic effects contributions produced by the knockdown of miR‐200c. CONCLUSION: Our studies suggest that miR‐200c may serve as a potential therapeutic target that could delay hypertrophy. We have also uncovered a relationship between miR‐200c and MLCK, identifying MLCK as a direct mediator of miR‐200c.
format Online
Article
Text
id pubmed-6433679
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-64336792019-04-08 Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway Hu, Shan Cheng, Mian Guo, Xin Wang, Shun Liu, Beilei Jiang, Hong Huang, Congxin Wu, Gang J Cell Mol Med Original Articles BACKGROUND: MicroRNAs (miRNAs) have been shown to commonly contribute to cardiac hypertrophy (CH). The aim of this study was to test the hypothesis that miR‐200c plays an important role in the progression of CH by targeting myosin light chain kinase (MLCK/MYLK). METHODS AND RESULTS: Cardiac hypertrophy was induced by aortic banding (AB) in rats. Cellular hypertrophy in neonatal rat cardiomyocytes (NCMs) was induced by AngII treatment. Echocardiography, histology and molecular measurements were used to assess the results of the experiments. The levels of apoptosis and reactive oxygen species (ROS) were also measured. Quantitative real‐time PCR (qRT‐PCR) and Western blotting were used to measure mRNA and protein levels respectively. The present results showed that miR‐200c expression was increased in response to CH both in vivo and in vitro. The down‐regulation of miRNA‐200c by a specific inhibitor markedly ameliorated CH resulting from AngII treatment, and the mRNA levels of atrial natriuretic peptide, brain natriuretic peptide and β‐myosin heavy chain were simultaneously decreased. Notably, minimal apoptosis and ROS accumulation were identified in AngII‐induced hypertrophic cardiomyocytes. Conversely, the up‐regulation of miR‐200c using specific mimics reversed these effects. Mechanistic investigations demonstrated that the MLCK gene is a direct target of miR‐200c; an increase in miR‐200c levels led to a decrease in the expression of MLCK and its downstream effector, p‐MLC2, while miR‐200c inhibition increased the expression of these proteins. Furthermore, inhibiting MLCK impaired the anti‐hypertrophic effects contributions produced by the knockdown of miR‐200c. CONCLUSION: Our studies suggest that miR‐200c may serve as a potential therapeutic target that could delay hypertrophy. We have also uncovered a relationship between miR‐200c and MLCK, identifying MLCK as a direct mediator of miR‐200c. John Wiley and Sons Inc. 2019-01-25 2019-04 /pmc/articles/PMC6433679/ /pubmed/30680929 http://dx.doi.org/10.1111/jcmm.14135 Text en © 2019 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine. This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Hu, Shan
Cheng, Mian
Guo, Xin
Wang, Shun
Liu, Beilei
Jiang, Hong
Huang, Congxin
Wu, Gang
Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title_full Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title_fullStr Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title_full_unstemmed Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title_short Down‐regulation of miR‐200c attenuates AngII‐induced cardiac hypertrophy via targeting the MLCK‐mediated pathway
title_sort down‐regulation of mir‐200c attenuates angii‐induced cardiac hypertrophy via targeting the mlck‐mediated pathway
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6433679/
https://www.ncbi.nlm.nih.gov/pubmed/30680929
http://dx.doi.org/10.1111/jcmm.14135
work_keys_str_mv AT hushan downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT chengmian downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT guoxin downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT wangshun downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT liubeilei downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT jianghong downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT huangcongxin downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway
AT wugang downregulationofmir200cattenuatesangiiinducedcardiachypertrophyviatargetingthemlckmediatedpathway