Cargando…

CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade

Multiple vaginal delivery (MVD) is an important factor for pelvic floor muscle (PFM) function decline and pelvic floor dysfunction (PFD). PFD is common in middle-aged and elderly women, but its pathogenesis is not clear. In this study, we found that the expression of CACNA1H was lower in the PFM of...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Suting, Hao, Menglei, Li, Bingshu, Chen, Mao, Chen, Jue, Tang, Jianming, Hong, Shasha, Min, Jie, Hu, Ming, Hong, Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7181873/
https://www.ncbi.nlm.nih.gov/pubmed/32332705
http://dx.doi.org/10.1038/s41419-020-2484-2
_version_ 1783526137656246272
author Li, Suting
Hao, Menglei
Li, Bingshu
Chen, Mao
Chen, Jue
Tang, Jianming
Hong, Shasha
Min, Jie
Hu, Ming
Hong, Li
author_facet Li, Suting
Hao, Menglei
Li, Bingshu
Chen, Mao
Chen, Jue
Tang, Jianming
Hong, Shasha
Min, Jie
Hu, Ming
Hong, Li
author_sort Li, Suting
collection PubMed
description Multiple vaginal delivery (MVD) is an important factor for pelvic floor muscle (PFM) function decline and pelvic floor dysfunction (PFD). PFD is common in middle-aged and elderly women, but its pathogenesis is not clear. In this study, we found that the expression of CACNA1H was lower in the PFM of old mice after MVD compared with old or adult mice. In in-vitro studies, we found that treatment with the T-type Ca(2+) channel (T-channel) inhibitor NNC-55 or downregulation of the CACNA1H gene by siRNA intervention promoted myotube atrophy and apoptosis. Mechanistically, we revealed that NNC-55 increased the expression of GRP78 and DDIT3 in myotubes, indicating endoplasmic reticulum stress (ERS) activation, and that the IRE1 and PERK pathways might be involved in this effect. NNC-55 induced the formation of autophagosomes but inhibited autophagy flux. Moreover, rapamycin, an autophagy activator, did not rescue myotube atrophy or apoptosis induced by NNC-55, and the autophagy inhibitors 3-MA and HCQ accelerated this damage. Further studies showed that the ERS inhibitors 4-PBA and TUDAC relieved NNC-55-induced damage and autophagy flux blockade. Finally, we found multisite muscle atrophy and decreased muscle function in Cacna1h(−/−) (TH-null) mice, as well as increased autophagy inhibition and apoptotic signals in the PFM of old WT mice after MVD and TH-null mice. Taken together, our results suggest that MVD-associated PFD is partially attributed to CACNA1H downregulation-induced PFM atrophy and that ERS is a potential therapeutic target for this disease.
format Online
Article
Text
id pubmed-7181873
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-71818732020-04-29 CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade Li, Suting Hao, Menglei Li, Bingshu Chen, Mao Chen, Jue Tang, Jianming Hong, Shasha Min, Jie Hu, Ming Hong, Li Cell Death Dis Article Multiple vaginal delivery (MVD) is an important factor for pelvic floor muscle (PFM) function decline and pelvic floor dysfunction (PFD). PFD is common in middle-aged and elderly women, but its pathogenesis is not clear. In this study, we found that the expression of CACNA1H was lower in the PFM of old mice after MVD compared with old or adult mice. In in-vitro studies, we found that treatment with the T-type Ca(2+) channel (T-channel) inhibitor NNC-55 or downregulation of the CACNA1H gene by siRNA intervention promoted myotube atrophy and apoptosis. Mechanistically, we revealed that NNC-55 increased the expression of GRP78 and DDIT3 in myotubes, indicating endoplasmic reticulum stress (ERS) activation, and that the IRE1 and PERK pathways might be involved in this effect. NNC-55 induced the formation of autophagosomes but inhibited autophagy flux. Moreover, rapamycin, an autophagy activator, did not rescue myotube atrophy or apoptosis induced by NNC-55, and the autophagy inhibitors 3-MA and HCQ accelerated this damage. Further studies showed that the ERS inhibitors 4-PBA and TUDAC relieved NNC-55-induced damage and autophagy flux blockade. Finally, we found multisite muscle atrophy and decreased muscle function in Cacna1h(−/−) (TH-null) mice, as well as increased autophagy inhibition and apoptotic signals in the PFM of old WT mice after MVD and TH-null mice. Taken together, our results suggest that MVD-associated PFD is partially attributed to CACNA1H downregulation-induced PFM atrophy and that ERS is a potential therapeutic target for this disease. Nature Publishing Group UK 2020-04-24 /pmc/articles/PMC7181873/ /pubmed/32332705 http://dx.doi.org/10.1038/s41419-020-2484-2 Text en © The Author(s) 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Li, Suting
Hao, Menglei
Li, Bingshu
Chen, Mao
Chen, Jue
Tang, Jianming
Hong, Shasha
Min, Jie
Hu, Ming
Hong, Li
CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title_full CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title_fullStr CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title_full_unstemmed CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title_short CACNA1H downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
title_sort cacna1h downregulation induces skeletal muscle atrophy involving endoplasmic reticulum stress activation and autophagy flux blockade
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7181873/
https://www.ncbi.nlm.nih.gov/pubmed/32332705
http://dx.doi.org/10.1038/s41419-020-2484-2
work_keys_str_mv AT lisuting cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT haomenglei cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT libingshu cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT chenmao cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT chenjue cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT tangjianming cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT hongshasha cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT minjie cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT huming cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade
AT hongli cacna1hdownregulationinducesskeletalmuscleatrophyinvolvingendoplasmicreticulumstressactivationandautophagyfluxblockade