Cargando…

HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver metabolic syndrome which affects millions of people worldwide. Recently, improving mitochondrial function and autophagic ability have been proposed as a means to prevent NAFLD. It has been previously described that high-temper...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Wei, Deng, Xueting, Zhu, Xiaolei, Yan, Qinhui, Zhou, Nan, Du, Susu, Li, Xiaonan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9023526/
https://www.ncbi.nlm.nih.gov/pubmed/35449197
http://dx.doi.org/10.1038/s41420-022-01022-4
_version_ 1784690371505160192
author Zhou, Wei
Deng, Xueting
Zhu, Xiaolei
Yan, Qinhui
Zhou, Nan
Du, Susu
Li, Xiaonan
author_facet Zhou, Wei
Deng, Xueting
Zhu, Xiaolei
Yan, Qinhui
Zhou, Nan
Du, Susu
Li, Xiaonan
author_sort Zhou, Wei
collection PubMed
description Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver metabolic syndrome which affects millions of people worldwide. Recently, improving mitochondrial function and autophagic ability have been proposed as a means to prevent NAFLD. It has been previously described that high-temperature requirement protein A2 (HtrA2/Omi) favors mitochondrial homeostasis and autophagy in hepatocytes. Thus, we explored the effects of HtrA2/Omi on regulating mitochondrial function and autophagy during NAFLD development. High-fat diet (HFD)-induced NAFLD in mice and free fatty acids (FFAs)-induced hepatocytes steatosis in vitro were established. Adeno-associated viruses (AAV) in vivo and plasmid in vitro were used to restore HtrA2/Omi expression. In this study, we reported that HtrA2/Omi expression considerably decreased in liver tissues from the HFD-induced NAFLD model and in L02 cells with FFA-treated. However, restoring HtrA2/Omi ameliorated hepatic steatosis, confirming by improved serum lipid profiles, glucose homeostasis, insulin resistance, histopathological lipid accumulation, and the gene expression related to lipid metabolism. Moreover, HtrA2/Omi also attenuated HFD-mediated mitochondrial dysfunction and autophagic blockage. TEM analysis revealed that liver mitochondrial structure and autophagosome formation were improved in hepatic HtrA2/Omi administration mice compared to HFD mice. And hepatic HtrA2/Omi overexpression enhanced mitochondrial fatty acid β-oxidation gene expression, elevated LC3II protein levels, induced LC3 puncta, and decreased SQSTM1/p62 protein levels. Furthermore, hepatic HtrA2/Omi increased respiratory exchange ratio and heat production in mice. Finally, HtrA2/Omi overexpression by plasmid significantly diminished lipid accumulation, mitochondrial dysfunction, and autophagic inhibition in FFA-treated L02 hepatocytes. Taken together, we demonstrated that HtrA2/Omi was a potential candidate for the treatment of NAFLD via improving mitochondrial functions, as well as restoring autophagic flux.
format Online
Article
Text
id pubmed-9023526
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-90235262022-04-28 HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux Zhou, Wei Deng, Xueting Zhu, Xiaolei Yan, Qinhui Zhou, Nan Du, Susu Li, Xiaonan Cell Death Discov Article Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver metabolic syndrome which affects millions of people worldwide. Recently, improving mitochondrial function and autophagic ability have been proposed as a means to prevent NAFLD. It has been previously described that high-temperature requirement protein A2 (HtrA2/Omi) favors mitochondrial homeostasis and autophagy in hepatocytes. Thus, we explored the effects of HtrA2/Omi on regulating mitochondrial function and autophagy during NAFLD development. High-fat diet (HFD)-induced NAFLD in mice and free fatty acids (FFAs)-induced hepatocytes steatosis in vitro were established. Adeno-associated viruses (AAV) in vivo and plasmid in vitro were used to restore HtrA2/Omi expression. In this study, we reported that HtrA2/Omi expression considerably decreased in liver tissues from the HFD-induced NAFLD model and in L02 cells with FFA-treated. However, restoring HtrA2/Omi ameliorated hepatic steatosis, confirming by improved serum lipid profiles, glucose homeostasis, insulin resistance, histopathological lipid accumulation, and the gene expression related to lipid metabolism. Moreover, HtrA2/Omi also attenuated HFD-mediated mitochondrial dysfunction and autophagic blockage. TEM analysis revealed that liver mitochondrial structure and autophagosome formation were improved in hepatic HtrA2/Omi administration mice compared to HFD mice. And hepatic HtrA2/Omi overexpression enhanced mitochondrial fatty acid β-oxidation gene expression, elevated LC3II protein levels, induced LC3 puncta, and decreased SQSTM1/p62 protein levels. Furthermore, hepatic HtrA2/Omi increased respiratory exchange ratio and heat production in mice. Finally, HtrA2/Omi overexpression by plasmid significantly diminished lipid accumulation, mitochondrial dysfunction, and autophagic inhibition in FFA-treated L02 hepatocytes. Taken together, we demonstrated that HtrA2/Omi was a potential candidate for the treatment of NAFLD via improving mitochondrial functions, as well as restoring autophagic flux. Nature Publishing Group UK 2022-04-21 /pmc/articles/PMC9023526/ /pubmed/35449197 http://dx.doi.org/10.1038/s41420-022-01022-4 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/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/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Zhou, Wei
Deng, Xueting
Zhu, Xiaolei
Yan, Qinhui
Zhou, Nan
Du, Susu
Li, Xiaonan
HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title_full HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title_fullStr HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title_full_unstemmed HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title_short HtrA2/Omi mitigates NAFLD in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
title_sort htra2/omi mitigates nafld in high-fat-fed mice by ameliorating mitochondrial dysfunction and restoring autophagic flux
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9023526/
https://www.ncbi.nlm.nih.gov/pubmed/35449197
http://dx.doi.org/10.1038/s41420-022-01022-4
work_keys_str_mv AT zhouwei htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT dengxueting htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT zhuxiaolei htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT yanqinhui htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT zhounan htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT dususu htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux
AT lixiaonan htra2omimitigatesnafldinhighfatfedmicebyamelioratingmitochondrialdysfunctionandrestoringautophagicflux