Cargando…

Modulation of adipose inflammation by cellular retinoic acid-binding protein 1

OBJECTIVES: Obesity, a metabolic syndrome, is known to be related to inflammation, especially adipose tissue inflammation. Cellular interactions within the expanded white adipose tissue (WAT) in obesity contribute to inflammation and studies have suggested that inflammation is triggered by inflamed...

Descripción completa

Detalles Bibliográficos
Autores principales: Wei, Chin-Wen, Nhieu, Jennifer, Lin, Yu-Lung, Wei, Li-Na
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9492549/
https://www.ncbi.nlm.nih.gov/pubmed/35794192
http://dx.doi.org/10.1038/s41366-022-01175-3
_version_ 1784793508253532160
author Wei, Chin-Wen
Nhieu, Jennifer
Lin, Yu-Lung
Wei, Li-Na
author_facet Wei, Chin-Wen
Nhieu, Jennifer
Lin, Yu-Lung
Wei, Li-Na
author_sort Wei, Chin-Wen
collection PubMed
description OBJECTIVES: Obesity, a metabolic syndrome, is known to be related to inflammation, especially adipose tissue inflammation. Cellular interactions within the expanded white adipose tissue (WAT) in obesity contribute to inflammation and studies have suggested that inflammation is triggered by inflamed adipocytes that recruit M1 macrophages into WAT. What causes accumulation of unhealthy adipocytes is an important topic of investigation. This study aims to understand the action of Cellular Retinoic Acid Binding Protein 1 (CRABP1) in WAT inflammation. METHODS: Eight weeks-old wild type (WT) and Crabp1 knockout (CKO) mice were fed with a normal diet (ND) or high-fat diet (HFD) for 8 weeks. Body weight and food intake were monitored. WATs and serum were collected for cellular and molecular analyses to determine affected signaling pathways. In cell culture studies, primary adipocyte differentiation and bone marrow-derived macrophages (BMDM) were used to examine adipocytes’ effects, mediated by CRABP1, in macrophage polarization. The 3T3L1-adipocyte was used to validate relevant signaling pathways. RESULTS: CKO mice developed an obese phenotype, more severely under high-fat diet (HFD) feeding. Further, CKO’s WAT exhibited a more severe inflammatory state as compared to wild type (WT) WAT, with a significantly expanded M1-like macrophage population. However, this was not caused by intrinsic defects of CKO macrophages. Rather, CKO adipocytes produced a significantly reduced level of adiponectin and had significantly lowered mitochondrial DNA content. CKO adipocyte-conditioned medium, compared to WT control, inhibited M2-like (CD206(+)) macrophage polarization. Mechanistically, defects in CKO adipocytes involved the ERK1/2 signaling pathway that could be modulated by CRABP1. CONCLUSIONS: This study shows that CRABP1 plays a protective role against HFD-induced WAT inflammation through, in part, its regulation of adiponectin production and mitochondrial homeostasis in adipocytes, thereby modulating macrophage polarization in WAT to control its inflammatory potential.
format Online
Article
Text
id pubmed-9492549
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-94925492022-09-23 Modulation of adipose inflammation by cellular retinoic acid-binding protein 1 Wei, Chin-Wen Nhieu, Jennifer Lin, Yu-Lung Wei, Li-Na Int J Obes (Lond) Article OBJECTIVES: Obesity, a metabolic syndrome, is known to be related to inflammation, especially adipose tissue inflammation. Cellular interactions within the expanded white adipose tissue (WAT) in obesity contribute to inflammation and studies have suggested that inflammation is triggered by inflamed adipocytes that recruit M1 macrophages into WAT. What causes accumulation of unhealthy adipocytes is an important topic of investigation. This study aims to understand the action of Cellular Retinoic Acid Binding Protein 1 (CRABP1) in WAT inflammation. METHODS: Eight weeks-old wild type (WT) and Crabp1 knockout (CKO) mice were fed with a normal diet (ND) or high-fat diet (HFD) for 8 weeks. Body weight and food intake were monitored. WATs and serum were collected for cellular and molecular analyses to determine affected signaling pathways. In cell culture studies, primary adipocyte differentiation and bone marrow-derived macrophages (BMDM) were used to examine adipocytes’ effects, mediated by CRABP1, in macrophage polarization. The 3T3L1-adipocyte was used to validate relevant signaling pathways. RESULTS: CKO mice developed an obese phenotype, more severely under high-fat diet (HFD) feeding. Further, CKO’s WAT exhibited a more severe inflammatory state as compared to wild type (WT) WAT, with a significantly expanded M1-like macrophage population. However, this was not caused by intrinsic defects of CKO macrophages. Rather, CKO adipocytes produced a significantly reduced level of adiponectin and had significantly lowered mitochondrial DNA content. CKO adipocyte-conditioned medium, compared to WT control, inhibited M2-like (CD206(+)) macrophage polarization. Mechanistically, defects in CKO adipocytes involved the ERK1/2 signaling pathway that could be modulated by CRABP1. CONCLUSIONS: This study shows that CRABP1 plays a protective role against HFD-induced WAT inflammation through, in part, its regulation of adiponectin production and mitochondrial homeostasis in adipocytes, thereby modulating macrophage polarization in WAT to control its inflammatory potential. Nature Publishing Group UK 2022-07-06 2022 /pmc/articles/PMC9492549/ /pubmed/35794192 http://dx.doi.org/10.1038/s41366-022-01175-3 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
Wei, Chin-Wen
Nhieu, Jennifer
Lin, Yu-Lung
Wei, Li-Na
Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title_full Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title_fullStr Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title_full_unstemmed Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title_short Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
title_sort modulation of adipose inflammation by cellular retinoic acid-binding protein 1
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9492549/
https://www.ncbi.nlm.nih.gov/pubmed/35794192
http://dx.doi.org/10.1038/s41366-022-01175-3
work_keys_str_mv AT weichinwen modulationofadiposeinflammationbycellularretinoicacidbindingprotein1
AT nhieujennifer modulationofadiposeinflammationbycellularretinoicacidbindingprotein1
AT linyulung modulationofadiposeinflammationbycellularretinoicacidbindingprotein1
AT weilina modulationofadiposeinflammationbycellularretinoicacidbindingprotein1