Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes

Abstract Background Ischemic diseases represent a major global health care burden. Angiogenesis is critical in recovery of blood flow and repair of injured tissue in ischemic diseases. Ten–eleven translocation protein 2 (TET2), a member of DNA demethylases, is involved in many pathological processes...

Full description

Bibliographic Details
Main Authors: Yefei Shi, Bo Li, Xinru Huang, Wenxin Kou, Ming Zhai, Yanxi Zeng, Shuangjie You, Qing Yu, Yifan Zhao, Jianhui Zhuang, Wenhui Peng, Weixia Jian
Format: Article
Language:English
Published: BMC 2023-01-01
Series:Cell & Bioscience
Subjects:
Online Access:https://doi.org/10.1186/s13578-023-00960-5
_version_ 1797945665900249088
author Yefei Shi
Bo Li
Xinru Huang
Wenxin Kou
Ming Zhai
Yanxi Zeng
Shuangjie You
Qing Yu
Yifan Zhao
Jianhui Zhuang
Wenhui Peng
Weixia Jian
author_facet Yefei Shi
Bo Li
Xinru Huang
Wenxin Kou
Ming Zhai
Yanxi Zeng
Shuangjie You
Qing Yu
Yifan Zhao
Jianhui Zhuang
Wenhui Peng
Weixia Jian
author_sort Yefei Shi
collection DOAJ
description Abstract Background Ischemic diseases represent a major global health care burden. Angiogenesis is critical in recovery of blood flow and repair of injured tissue in ischemic diseases. Ten–eleven translocation protein 2 (TET2), a member of DNA demethylases, is involved in many pathological processes. However, the role of TET2 in angiogenesis is still unrevealed. Methods TET2 was screened out from three DNA demethylases involved in 5-hydroxylmethylcytosine (5-hmC) regulation, including TET1, TET2 and TET3. Knockdown by small interfering RNAs and overexpression by adenovirus were used to evaluate the role of TET2 on the function of endothelial cells. The blood flow recovery and density of capillary were analyzed in the endothelial cells-specific TET2-deficient mice. RNA sequencing was used to identify the TET2-mediated mechanisms under hypoxia. Co-immunoprecipitation (Co-IP), chromatin immunoprecipitation-qPCR (ChIP-qPCR) and glucosylated hydroxymethyl-sensitive-qPCR (GluMS-qPCR) were further performed to reveal the interaction of TET2 and STAT3. Results TET2 was significantly downregulated in endothelial cells under hypoxia and led to a global decrease of 5-hmC level. TET2 knockdown aggravated the hypoxia‐induced dysfunction of endothelial cells, while TET2 overexpression alleviated the hypoxia‐induced dysfunction. Meanwhile, the deficiency of TET2 in endothelial cells impaired blood flow recovery and the density of capillary in the mouse model of hindlimb ischemia. Mechanistically, RNA sequencing indicated that the STAT3 signaling pathway was significantly inhibited by TET2 knockdown. Additionally, Co-IP, ChIP-qPCR and GluMS-qPCR further illustrated that STAT3 recruited and physically interacted with TET2 to activate STAT3 target genes. As expected, the effects of TET2 overexpression were completely suppressed by STAT3 silencing in vitro. Conclusions Our study suggests that the deficiency of TET2 in endothelial cells impairs angiogenesis via suppression of the STAT3 signaling pathway. These findings give solid evidence for TET2 to be a therapeutic alternative for ischemic diseases. Graphical Abstract
first_indexed 2024-04-10T20:59:46Z
format Article
id doaj.art-4948448a344949c9b697309fc5d3b455
institution Directory Open Access Journal
issn 2045-3701
language English
last_indexed 2024-04-10T20:59:46Z
publishDate 2023-01-01
publisher BMC
record_format Article
series Cell & Bioscience
spelling doaj.art-4948448a344949c9b697309fc5d3b4552023-01-22T12:26:19ZengBMCCell & Bioscience2045-37012023-01-0113111610.1186/s13578-023-00960-5Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genesYefei Shi0Bo Li1Xinru Huang2Wenxin Kou3Ming Zhai4Yanxi Zeng5Shuangjie You6Qing Yu7Yifan Zhao8Jianhui Zhuang9Wenhui Peng10Weixia Jian11Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of MedicineDepartment of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of MedicineAbstract Background Ischemic diseases represent a major global health care burden. Angiogenesis is critical in recovery of blood flow and repair of injured tissue in ischemic diseases. Ten–eleven translocation protein 2 (TET2), a member of DNA demethylases, is involved in many pathological processes. However, the role of TET2 in angiogenesis is still unrevealed. Methods TET2 was screened out from three DNA demethylases involved in 5-hydroxylmethylcytosine (5-hmC) regulation, including TET1, TET2 and TET3. Knockdown by small interfering RNAs and overexpression by adenovirus were used to evaluate the role of TET2 on the function of endothelial cells. The blood flow recovery and density of capillary were analyzed in the endothelial cells-specific TET2-deficient mice. RNA sequencing was used to identify the TET2-mediated mechanisms under hypoxia. Co-immunoprecipitation (Co-IP), chromatin immunoprecipitation-qPCR (ChIP-qPCR) and glucosylated hydroxymethyl-sensitive-qPCR (GluMS-qPCR) were further performed to reveal the interaction of TET2 and STAT3. Results TET2 was significantly downregulated in endothelial cells under hypoxia and led to a global decrease of 5-hmC level. TET2 knockdown aggravated the hypoxia‐induced dysfunction of endothelial cells, while TET2 overexpression alleviated the hypoxia‐induced dysfunction. Meanwhile, the deficiency of TET2 in endothelial cells impaired blood flow recovery and the density of capillary in the mouse model of hindlimb ischemia. Mechanistically, RNA sequencing indicated that the STAT3 signaling pathway was significantly inhibited by TET2 knockdown. Additionally, Co-IP, ChIP-qPCR and GluMS-qPCR further illustrated that STAT3 recruited and physically interacted with TET2 to activate STAT3 target genes. As expected, the effects of TET2 overexpression were completely suppressed by STAT3 silencing in vitro. Conclusions Our study suggests that the deficiency of TET2 in endothelial cells impairs angiogenesis via suppression of the STAT3 signaling pathway. These findings give solid evidence for TET2 to be a therapeutic alternative for ischemic diseases. Graphical Abstracthttps://doi.org/10.1186/s13578-023-00960-5TET25-hmCSTAT3AngiogenesisIschemic diseases
spellingShingle Yefei Shi
Bo Li
Xinru Huang
Wenxin Kou
Ming Zhai
Yanxi Zeng
Shuangjie You
Qing Yu
Yifan Zhao
Jianhui Zhuang
Wenhui Peng
Weixia Jian
Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
Cell & Bioscience
TET2
5-hmC
STAT3
Angiogenesis
Ischemic diseases
title Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
title_full Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
title_fullStr Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
title_full_unstemmed Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
title_short Loss of TET2 impairs endothelial angiogenesis via downregulating STAT3 target genes
title_sort loss of tet2 impairs endothelial angiogenesis via downregulating stat3 target genes
topic TET2
5-hmC
STAT3
Angiogenesis
Ischemic diseases
url https://doi.org/10.1186/s13578-023-00960-5
work_keys_str_mv AT yefeishi lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT boli lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT xinruhuang lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT wenxinkou lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT mingzhai lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT yanxizeng lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT shuangjieyou lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT qingyu lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT yifanzhao lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT jianhuizhuang lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT wenhuipeng lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes
AT weixiajian lossoftet2impairsendothelialangiogenesisviadownregulatingstat3targetgenes