Cargando…

MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1

BACKGROUND: A critical mechanism in atrial fibrillation (AF) is cardiac autonomic nerve remodeling (ANR). MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the post-transcriptional level. Numerous miRNAs are involved in diseases of the nervous and cardiovascular systems....

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yujiao, Zheng, Shaohua, Geng, Yangyang, Xue, Jiao, Wang, Zhongsu, Xie, Xinxing, Wang, Jiangrong, Zhang, Shuyu, Hou, Yinglong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4376950/
https://www.ncbi.nlm.nih.gov/pubmed/25816284
http://dx.doi.org/10.1371/journal.pone.0122674
_version_ 1782363819243208704
author Zhang, Yujiao
Zheng, Shaohua
Geng, Yangyang
Xue, Jiao
Wang, Zhongsu
Xie, Xinxing
Wang, Jiangrong
Zhang, Shuyu
Hou, Yinglong
author_facet Zhang, Yujiao
Zheng, Shaohua
Geng, Yangyang
Xue, Jiao
Wang, Zhongsu
Xie, Xinxing
Wang, Jiangrong
Zhang, Shuyu
Hou, Yinglong
author_sort Zhang, Yujiao
collection PubMed
description BACKGROUND: A critical mechanism in atrial fibrillation (AF) is cardiac autonomic nerve remodeling (ANR). MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the post-transcriptional level. Numerous miRNAs are involved in diseases of the nervous and cardiovascular systems. OBJECTIVE: We aimed to assess the underlying role of miRNAs in regulating cardiac ANR in AF by right atrial tachypacing (A-TP) in canines. METHODS AND RESULTS: Following 4-week A-TP, the superior left ganglionated plexuses (SLGPs), which are embedded in the fat pads of the left atrium, were subjected to miRNA expression profiling to screen preferentially expressed miRNAs. Sixteen miRNAs showed significantly differential expression between the control and A-TP groups, including miR-206, miR-203, miR-224 and miR-137. In particular, we focused on miR-206, which was elevated ~10-fold in A-TP dogs. Forced expression of miR-206 through lentiviral infection based on A-TP in vivo significantly shortened the atrial effective refractory period (AERP) (81 ± 7 vs. 98 ± 7 ms, P < 0.05). Immunohistochemical analysis showed that the regeneration of nerves increased more than 2-fold by miR-206 overexpression (P < 0.01). The expression of superoxide dismutase 1 (SOD1) was repressed by miR-206 overexpression by Western blot and luciferase assay, indicative of SOD1 as a direct target of miR-206. Overexpression of miR-206 increased reactive oxygen species (ROS) levels in vitro and in vivo, whereas miR-206 silencing attenuated irradiation- or A-TP-induced ROS. Knockdown of SOD1 effectively abolished ROS reduction caused by miR-206 silencing. CONCLUSIONS: Our results found the differential expression of miRNAs in response to ANR in AF and elucidated the important role of miR-206 by targeting SOD1. The study illustrated the novel molecular mechanism of ANR and indicated a potential therapeutic target for AF.
format Online
Article
Text
id pubmed-4376950
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-43769502015-04-04 MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1 Zhang, Yujiao Zheng, Shaohua Geng, Yangyang Xue, Jiao Wang, Zhongsu Xie, Xinxing Wang, Jiangrong Zhang, Shuyu Hou, Yinglong PLoS One Research Article BACKGROUND: A critical mechanism in atrial fibrillation (AF) is cardiac autonomic nerve remodeling (ANR). MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the post-transcriptional level. Numerous miRNAs are involved in diseases of the nervous and cardiovascular systems. OBJECTIVE: We aimed to assess the underlying role of miRNAs in regulating cardiac ANR in AF by right atrial tachypacing (A-TP) in canines. METHODS AND RESULTS: Following 4-week A-TP, the superior left ganglionated plexuses (SLGPs), which are embedded in the fat pads of the left atrium, were subjected to miRNA expression profiling to screen preferentially expressed miRNAs. Sixteen miRNAs showed significantly differential expression between the control and A-TP groups, including miR-206, miR-203, miR-224 and miR-137. In particular, we focused on miR-206, which was elevated ~10-fold in A-TP dogs. Forced expression of miR-206 through lentiviral infection based on A-TP in vivo significantly shortened the atrial effective refractory period (AERP) (81 ± 7 vs. 98 ± 7 ms, P < 0.05). Immunohistochemical analysis showed that the regeneration of nerves increased more than 2-fold by miR-206 overexpression (P < 0.01). The expression of superoxide dismutase 1 (SOD1) was repressed by miR-206 overexpression by Western blot and luciferase assay, indicative of SOD1 as a direct target of miR-206. Overexpression of miR-206 increased reactive oxygen species (ROS) levels in vitro and in vivo, whereas miR-206 silencing attenuated irradiation- or A-TP-induced ROS. Knockdown of SOD1 effectively abolished ROS reduction caused by miR-206 silencing. CONCLUSIONS: Our results found the differential expression of miRNAs in response to ANR in AF and elucidated the important role of miR-206 by targeting SOD1. The study illustrated the novel molecular mechanism of ANR and indicated a potential therapeutic target for AF. Public Library of Science 2015-03-27 /pmc/articles/PMC4376950/ /pubmed/25816284 http://dx.doi.org/10.1371/journal.pone.0122674 Text en © 2015 Zhang et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Zhang, Yujiao
Zheng, Shaohua
Geng, Yangyang
Xue, Jiao
Wang, Zhongsu
Xie, Xinxing
Wang, Jiangrong
Zhang, Shuyu
Hou, Yinglong
MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title_full MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title_fullStr MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title_full_unstemmed MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title_short MicroRNA Profiling of Atrial Fibrillation in Canines: MiR-206 Modulates Intrinsic Cardiac Autonomic Nerve Remodeling by Regulating SOD1
title_sort microrna profiling of atrial fibrillation in canines: mir-206 modulates intrinsic cardiac autonomic nerve remodeling by regulating sod1
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4376950/
https://www.ncbi.nlm.nih.gov/pubmed/25816284
http://dx.doi.org/10.1371/journal.pone.0122674
work_keys_str_mv AT zhangyujiao micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT zhengshaohua micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT gengyangyang micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT xuejiao micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT wangzhongsu micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT xiexinxing micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT wangjiangrong micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT zhangshuyu micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1
AT houyinglong micrornaprofilingofatrialfibrillationincaninesmir206modulatesintrinsiccardiacautonomicnerveremodelingbyregulatingsod1