Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans

IntroductionVascular dysfunction and chronic inflammation are characteristics of obesity-induced adipose tissue dysfunction. Proinflammatory cytokines can drive an endothelial-to-mesenchymal transition (EndoMT), where endothelial cells undergo a phenotypic switch to mesenchymal-like cells that are p...

Full description

Bibliographic Details
Main Authors: Nicholas W. Chavkin, Tanvi Vippa, Changhee Jung, Stephanie McDonnell, Karen K. Hirschi, Noyan Gokce, Kenneth Walsh
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-09-01
Series:Frontiers in Cardiovascular Medicine
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcvm.2023.1264479/full
_version_ 1797680814432976896
author Nicholas W. Chavkin
Nicholas W. Chavkin
Tanvi Vippa
Changhee Jung
Stephanie McDonnell
Karen K. Hirschi
Karen K. Hirschi
Karen K. Hirschi
Noyan Gokce
Kenneth Walsh
Kenneth Walsh
author_facet Nicholas W. Chavkin
Nicholas W. Chavkin
Tanvi Vippa
Changhee Jung
Stephanie McDonnell
Karen K. Hirschi
Karen K. Hirschi
Karen K. Hirschi
Noyan Gokce
Kenneth Walsh
Kenneth Walsh
author_sort Nicholas W. Chavkin
collection DOAJ
description IntroductionVascular dysfunction and chronic inflammation are characteristics of obesity-induced adipose tissue dysfunction. Proinflammatory cytokines can drive an endothelial-to-mesenchymal transition (EndoMT), where endothelial cells undergo a phenotypic switch to mesenchymal-like cells that are pro-inflammatory and pro-fibrotic. In this study, we sought to determine whether obesity can promote EndoMT in adipose tissue.MethodsMice in which endothelial cells are lineage-traced with eYFP were fed a high-fat/high-sucrose (HF/HS) or Control diet for 13, 26, and 52 weeks, and EndoMT was assessed in adipose tissue depots as percentage of CD45−CD31−Acta2+ mesenchymal-like cells that were eYFP +. EndoMT was also assessed in human adipose endothelial cells through cell culture assays and by the analysis of single cell RNA sequencing datasets obtained from the visceral adipose tissues of obese individuals.ResultsQuantification by flow cytometry showed that mice fed a HF/HS diet display a time-dependent increase in EndoMT over Control diet in subcutaneous adipose tissue (+3.0%, +2.6-fold at 13 weeks; +10.6%, +3.2-fold at 26 weeks; +11.8%, +2.9-fold at 52 weeks) and visceral adipose tissue (+5.5%, +2.3-fold at 13 weeks; +20.7%, +4.3-fold at 26 weeks; +25.7%, +4.8-fold at 52 weeks). Transcriptomic analysis revealed that EndoMT cells in visceral adipose tissue have enriched expression of genes associated with inflammatory and TGFβ signaling pathways. Human adipose-derived microvascular endothelial cells cultured with TGF-β1, IFN-γ, and TNF-α exhibited a similar upregulation of EndoMT markers and induction of inflammatory response pathways. Analysis of single cell RNA sequencing datasets from visceral adipose tissue of obese patients revealed a nascent EndoMT sub-cluster of endothelial cells with reduced PECAM1 and increased ACTA2 expression, which was also enriched for inflammatory signaling genes and other genes associated with EndoMT.DiscussionThese experimental and clinical findings show that chronic obesity can accelerate EndoMT in adipose tissue. We speculate that EndoMT is a feature of adipose tissue dysfunction that contributes to local inflammation and the systemic metabolic effects of obesity..
first_indexed 2024-03-11T23:35:44Z
format Article
id doaj.art-27e90ddeacdf4a618020467162ef7cc4
institution Directory Open Access Journal
issn 2297-055X
language English
last_indexed 2024-03-11T23:35:44Z
publishDate 2023-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cardiovascular Medicine
spelling doaj.art-27e90ddeacdf4a618020467162ef7cc42023-09-20T03:16:08ZengFrontiers Media S.A.Frontiers in Cardiovascular Medicine2297-055X2023-09-011010.3389/fcvm.2023.12644791264479Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humansNicholas W. Chavkin0Nicholas W. Chavkin1Tanvi Vippa2Changhee Jung3Stephanie McDonnell4Karen K. Hirschi5Karen K. Hirschi6Karen K. Hirschi7Noyan Gokce8Kenneth Walsh9Kenneth Walsh10Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesDepartment of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United StatesRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesDepartment of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United StatesRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesDepartment of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United StatesDepartment of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, United StatesDepartment of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, United StatesRobert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesHematovascular Biology Center, University of Virginia School of Medicine, Charlottesville, VA, United StatesIntroductionVascular dysfunction and chronic inflammation are characteristics of obesity-induced adipose tissue dysfunction. Proinflammatory cytokines can drive an endothelial-to-mesenchymal transition (EndoMT), where endothelial cells undergo a phenotypic switch to mesenchymal-like cells that are pro-inflammatory and pro-fibrotic. In this study, we sought to determine whether obesity can promote EndoMT in adipose tissue.MethodsMice in which endothelial cells are lineage-traced with eYFP were fed a high-fat/high-sucrose (HF/HS) or Control diet for 13, 26, and 52 weeks, and EndoMT was assessed in adipose tissue depots as percentage of CD45−CD31−Acta2+ mesenchymal-like cells that were eYFP +. EndoMT was also assessed in human adipose endothelial cells through cell culture assays and by the analysis of single cell RNA sequencing datasets obtained from the visceral adipose tissues of obese individuals.ResultsQuantification by flow cytometry showed that mice fed a HF/HS diet display a time-dependent increase in EndoMT over Control diet in subcutaneous adipose tissue (+3.0%, +2.6-fold at 13 weeks; +10.6%, +3.2-fold at 26 weeks; +11.8%, +2.9-fold at 52 weeks) and visceral adipose tissue (+5.5%, +2.3-fold at 13 weeks; +20.7%, +4.3-fold at 26 weeks; +25.7%, +4.8-fold at 52 weeks). Transcriptomic analysis revealed that EndoMT cells in visceral adipose tissue have enriched expression of genes associated with inflammatory and TGFβ signaling pathways. Human adipose-derived microvascular endothelial cells cultured with TGF-β1, IFN-γ, and TNF-α exhibited a similar upregulation of EndoMT markers and induction of inflammatory response pathways. Analysis of single cell RNA sequencing datasets from visceral adipose tissue of obese patients revealed a nascent EndoMT sub-cluster of endothelial cells with reduced PECAM1 and increased ACTA2 expression, which was also enriched for inflammatory signaling genes and other genes associated with EndoMT.DiscussionThese experimental and clinical findings show that chronic obesity can accelerate EndoMT in adipose tissue. We speculate that EndoMT is a feature of adipose tissue dysfunction that contributes to local inflammation and the systemic metabolic effects of obesity..https://www.frontiersin.org/articles/10.3389/fcvm.2023.1264479/fullendothelial-to-mesenchymal transitionobesityadiposeendotheliumagingvascular biology
spellingShingle Nicholas W. Chavkin
Nicholas W. Chavkin
Tanvi Vippa
Changhee Jung
Stephanie McDonnell
Karen K. Hirschi
Karen K. Hirschi
Karen K. Hirschi
Noyan Gokce
Kenneth Walsh
Kenneth Walsh
Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
Frontiers in Cardiovascular Medicine
endothelial-to-mesenchymal transition
obesity
adipose
endothelium
aging
vascular biology
title Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
title_full Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
title_fullStr Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
title_full_unstemmed Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
title_short Obesity accelerates endothelial-to-mesenchymal transition in adipose tissues of mice and humans
title_sort obesity accelerates endothelial to mesenchymal transition in adipose tissues of mice and humans
topic endothelial-to-mesenchymal transition
obesity
adipose
endothelium
aging
vascular biology
url https://www.frontiersin.org/articles/10.3389/fcvm.2023.1264479/full
work_keys_str_mv AT nicholaswchavkin obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT nicholaswchavkin obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT tanvivippa obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT changheejung obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT stephaniemcdonnell obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT karenkhirschi obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT karenkhirschi obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT karenkhirschi obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT noyangokce obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT kennethwalsh obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans
AT kennethwalsh obesityacceleratesendothelialtomesenchymaltransitioninadiposetissuesofmiceandhumans