Cargando…

ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis

Obesity has become an extensive threat to human health due to associated chronic inflammation and metabolic diseases. Apoptosis-associated speck-like protein (ASC) is a critical link between inflammasome and apoptosis-inducing proteins. In this study, we aimed to clarify the role of ASC in lipid met...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Hong, Pei, Qilin, Tao, Linfen, Xia, Jing, Lu, Guocai, Zong, Ying, Xie, Wenhua, Li, Wanqing, Huang, Chenglong, Zeng, Ting, Yu, Xinyu, Wang, Weixuan, Chen, Gaojun, Yang, Song, Cheng, Rui, Li, Xi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9456541/
https://www.ncbi.nlm.nih.gov/pubmed/36077447
http://dx.doi.org/10.3390/ijms231710042
_version_ 1784785840533143552
author Chen, Hong
Pei, Qilin
Tao, Linfen
Xia, Jing
Lu, Guocai
Zong, Ying
Xie, Wenhua
Li, Wanqing
Huang, Chenglong
Zeng, Ting
Yu, Xinyu
Wang, Weixuan
Chen, Gaojun
Yang, Song
Cheng, Rui
Li, Xi
author_facet Chen, Hong
Pei, Qilin
Tao, Linfen
Xia, Jing
Lu, Guocai
Zong, Ying
Xie, Wenhua
Li, Wanqing
Huang, Chenglong
Zeng, Ting
Yu, Xinyu
Wang, Weixuan
Chen, Gaojun
Yang, Song
Cheng, Rui
Li, Xi
author_sort Chen, Hong
collection PubMed
description Obesity has become an extensive threat to human health due to associated chronic inflammation and metabolic diseases. Apoptosis-associated speck-like protein (ASC) is a critical link between inflammasome and apoptosis-inducing proteins. In this study, we aimed to clarify the role of ASC in lipid metabolism. With high-fat diet (HFD) and knockout leptin gene mice (ob/ob), we found that ASC expression in subcutaneous adipose tissue (SAT) correlated with obesity. It could also positively regulate the reprogramming of cellular energy metabolism. Stromal vascular fractions (SVF) cells derived from the SAT of Asc(−/−) mice or SVF from wild-type (WT) mice transfected with ASC siRNA were used to further investigate the underlying molecular mechanisms. We found ASC deficiency could lead to lipogenesis and inhibit lipolysis in SAT, aggravating lipid accumulation and impairing metabolic balance. In addition, our results showed that p53 and AMPKα expression were inhibited in SAT when ASC level was low. p53 and AMP-activated protein kinase α (AMPKα) were then assessed to elucidate whether they were downstream of ASC in regulating lipid metabolism. Our results revealed that ASC deficiency could promote lipid accumulation by increasing lipogenesis and decreasing lipolysis through p53/AMPKα axis. Regulation of ASC on lipid metabolism might be a novel therapeutic target for obesity.
format Online
Article
Text
id pubmed-9456541
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-94565412022-09-09 ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis Chen, Hong Pei, Qilin Tao, Linfen Xia, Jing Lu, Guocai Zong, Ying Xie, Wenhua Li, Wanqing Huang, Chenglong Zeng, Ting Yu, Xinyu Wang, Weixuan Chen, Gaojun Yang, Song Cheng, Rui Li, Xi Int J Mol Sci Article Obesity has become an extensive threat to human health due to associated chronic inflammation and metabolic diseases. Apoptosis-associated speck-like protein (ASC) is a critical link between inflammasome and apoptosis-inducing proteins. In this study, we aimed to clarify the role of ASC in lipid metabolism. With high-fat diet (HFD) and knockout leptin gene mice (ob/ob), we found that ASC expression in subcutaneous adipose tissue (SAT) correlated with obesity. It could also positively regulate the reprogramming of cellular energy metabolism. Stromal vascular fractions (SVF) cells derived from the SAT of Asc(−/−) mice or SVF from wild-type (WT) mice transfected with ASC siRNA were used to further investigate the underlying molecular mechanisms. We found ASC deficiency could lead to lipogenesis and inhibit lipolysis in SAT, aggravating lipid accumulation and impairing metabolic balance. In addition, our results showed that p53 and AMPKα expression were inhibited in SAT when ASC level was low. p53 and AMP-activated protein kinase α (AMPKα) were then assessed to elucidate whether they were downstream of ASC in regulating lipid metabolism. Our results revealed that ASC deficiency could promote lipid accumulation by increasing lipogenesis and decreasing lipolysis through p53/AMPKα axis. Regulation of ASC on lipid metabolism might be a novel therapeutic target for obesity. MDPI 2022-09-02 /pmc/articles/PMC9456541/ /pubmed/36077447 http://dx.doi.org/10.3390/ijms231710042 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Chen, Hong
Pei, Qilin
Tao, Linfen
Xia, Jing
Lu, Guocai
Zong, Ying
Xie, Wenhua
Li, Wanqing
Huang, Chenglong
Zeng, Ting
Yu, Xinyu
Wang, Weixuan
Chen, Gaojun
Yang, Song
Cheng, Rui
Li, Xi
ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title_full ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title_fullStr ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title_full_unstemmed ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title_short ASC Regulates Subcutaneous Adipose Tissue Lipogenesis and Lipolysis via p53/AMPKα Axis
title_sort asc regulates subcutaneous adipose tissue lipogenesis and lipolysis via p53/ampkα axis
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9456541/
https://www.ncbi.nlm.nih.gov/pubmed/36077447
http://dx.doi.org/10.3390/ijms231710042
work_keys_str_mv AT chenhong ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT peiqilin ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT taolinfen ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT xiajing ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT luguocai ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT zongying ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT xiewenhua ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT liwanqing ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT huangchenglong ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT zengting ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT yuxinyu ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT wangweixuan ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT chengaojun ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT yangsong ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT chengrui ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis
AT lixi ascregulatessubcutaneousadiposetissuelipogenesisandlipolysisviap53ampkaaxis