LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization

Aims: Metabolic switching during heart development contributes to postnatal cardiomyocyte (CM) cell cycle exit and loss of regenerative capacity in the mammalian heart. Metabolic control has potential for developing effective CM proliferation strategies. We sought to determine whether lactate dehydr...

Full description

Bibliographic Details
Main Authors: Yijin Chen, Guangkai Wu, Mengsha Li, Michael Hesse, Yusheng Ma, Wei Chen, Haoxiang Huang, Yu Liu, Wenlong Xu, Yating Tang, Hao Zheng, Chuling Li, Zhongqiu Lin, Guojun Chen, Wangjun Liao, Yulin Liao, Jianping Bin, Yanmei Chen
Format: Article
Language:English
Published: Elsevier 2022-10-01
Series:Redox Biology
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S221323172200218X
_version_ 1811267026334777344
author Yijin Chen
Guangkai Wu
Mengsha Li
Michael Hesse
Yusheng Ma
Wei Chen
Haoxiang Huang
Yu Liu
Wenlong Xu
Yating Tang
Hao Zheng
Chuling Li
Zhongqiu Lin
Guojun Chen
Wangjun Liao
Yulin Liao
Jianping Bin
Yanmei Chen
author_facet Yijin Chen
Guangkai Wu
Mengsha Li
Michael Hesse
Yusheng Ma
Wei Chen
Haoxiang Huang
Yu Liu
Wenlong Xu
Yating Tang
Hao Zheng
Chuling Li
Zhongqiu Lin
Guojun Chen
Wangjun Liao
Yulin Liao
Jianping Bin
Yanmei Chen
author_sort Yijin Chen
collection DOAJ
description Aims: Metabolic switching during heart development contributes to postnatal cardiomyocyte (CM) cell cycle exit and loss of regenerative capacity in the mammalian heart. Metabolic control has potential for developing effective CM proliferation strategies. We sought to determine whether lactate dehydrogenase A (LDHA) regulated CM proliferation by inducing metabolic reprogramming. Methods and results: LDHA expression was high in P1 hearts and significantly decreased during postnatal heart development. CM-specific LDHA knockout mice were generated using CRISPR/Cas9 technology. CM-specific LDHA knockout inhibited CM proliferation, leading to worse cardiac function and a lower survival rate in the neonatal apical resection model. In contrast, CM-specific overexpression of LDHA promoted CM proliferation and cardiac repair post-MI. The α-MHC-H2B-mCh/CAG-eGFP-anillin system was used to confirm the proliferative effect triggered by LDHA on P7 CMs and adult hearts. Metabolomics, proteomics and Co-IP experiments indicated that LDHA-mediated succinyl coenzyme A reduction inhibited succinylation-dependent ubiquitination of thioredoxin reductase 1 (Txnrd1), which alleviated ROS and thereby promoted CM proliferation. In addition, flow cytometry and western blotting showed that LDHA-driven lactate production created a beneficial cardiac regenerative microenvironment by inducing M2 macrophage polarization. Conclusions: LDHA-mediated metabolic reprogramming promoted CM proliferation by alleviating ROS and inducing M2 macrophage polarization, indicating that LDHA might be an effective target for promoting cardiac repair post-MI.
first_indexed 2024-04-12T20:53:57Z
format Article
id doaj.art-cbea050ec87f401b9a5a184a2000bb60
institution Directory Open Access Journal
issn 2213-2317
language English
last_indexed 2024-04-12T20:53:57Z
publishDate 2022-10-01
publisher Elsevier
record_format Article
series Redox Biology
spelling doaj.art-cbea050ec87f401b9a5a184a2000bb602022-12-22T03:17:02ZengElsevierRedox Biology2213-23172022-10-0156102446LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarizationYijin Chen0Guangkai Wu1Mengsha Li2Michael Hesse3Yusheng Ma4Wei Chen5Haoxiang Huang6Yu Liu7Wenlong Xu8Yating Tang9Hao Zheng10Chuling Li11Zhongqiu Lin12Guojun Chen13Wangjun Liao14Yulin Liao15Jianping Bin16Yanmei Chen17Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, China; Guizhou University Hospital, Guiyang Guizhou, 550025, ChinaInstitute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, GermanyDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, ChinaDepartment of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, China; Corresponding author. Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China.Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510515, Guangzhou, China; Corresponding author. Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China.Aims: Metabolic switching during heart development contributes to postnatal cardiomyocyte (CM) cell cycle exit and loss of regenerative capacity in the mammalian heart. Metabolic control has potential for developing effective CM proliferation strategies. We sought to determine whether lactate dehydrogenase A (LDHA) regulated CM proliferation by inducing metabolic reprogramming. Methods and results: LDHA expression was high in P1 hearts and significantly decreased during postnatal heart development. CM-specific LDHA knockout mice were generated using CRISPR/Cas9 technology. CM-specific LDHA knockout inhibited CM proliferation, leading to worse cardiac function and a lower survival rate in the neonatal apical resection model. In contrast, CM-specific overexpression of LDHA promoted CM proliferation and cardiac repair post-MI. The α-MHC-H2B-mCh/CAG-eGFP-anillin system was used to confirm the proliferative effect triggered by LDHA on P7 CMs and adult hearts. Metabolomics, proteomics and Co-IP experiments indicated that LDHA-mediated succinyl coenzyme A reduction inhibited succinylation-dependent ubiquitination of thioredoxin reductase 1 (Txnrd1), which alleviated ROS and thereby promoted CM proliferation. In addition, flow cytometry and western blotting showed that LDHA-driven lactate production created a beneficial cardiac regenerative microenvironment by inducing M2 macrophage polarization. Conclusions: LDHA-mediated metabolic reprogramming promoted CM proliferation by alleviating ROS and inducing M2 macrophage polarization, indicating that LDHA might be an effective target for promoting cardiac repair post-MI.http://www.sciencedirect.com/science/article/pii/S221323172200218XLDHAMetabolic reprogrammingCardiomyocyte proliferationROSMacrophage polarization
spellingShingle Yijin Chen
Guangkai Wu
Mengsha Li
Michael Hesse
Yusheng Ma
Wei Chen
Haoxiang Huang
Yu Liu
Wenlong Xu
Yating Tang
Hao Zheng
Chuling Li
Zhongqiu Lin
Guojun Chen
Wangjun Liao
Yulin Liao
Jianping Bin
Yanmei Chen
LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
Redox Biology
LDHA
Metabolic reprogramming
Cardiomyocyte proliferation
ROS
Macrophage polarization
title LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
title_full LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
title_fullStr LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
title_full_unstemmed LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
title_short LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization
title_sort ldha mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ros and inducing m2 macrophage polarization
topic LDHA
Metabolic reprogramming
Cardiomyocyte proliferation
ROS
Macrophage polarization
url http://www.sciencedirect.com/science/article/pii/S221323172200218X
work_keys_str_mv AT yijinchen ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT guangkaiwu ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT mengshali ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT michaelhesse ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT yushengma ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT weichen ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT haoxianghuang ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT yuliu ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT wenlongxu ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT yatingtang ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT haozheng ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT chulingli ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT zhongqiulin ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT guojunchen ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT wangjunliao ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT yulinliao ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT jianpingbin ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization
AT yanmeichen ldhamediatedmetabolicreprogrammingpromotedcardiomyocyteproliferationbyalleviatingrosandinducingm2macrophagepolarization