CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions

Induced autophagy is protective against myocardial hypoxia/ischemia (H/I) injury, but evidence regarding the extent of autophagic clearance under H/I and the molecular mechanisms that influence autophagic flux has scarcely been presented. Here, we report that CD38 knockout improved cardiac function...

Full description

Bibliographic Details
Main Authors: Xingyue Zhang, Lingfei Li, Qiong Zhang, Qinglin Wei, Jiezhi Lin, Jiezhi Jia, Junhui Zhang, Tiantian Yan, Yanling Lv, Xupin Jiang, Peng Zhang, Huapei Song, Dongxia Zhang, Yuesheng Huang
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-04-01
Series:Frontiers in Cell and Developmental Biology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fcell.2020.00191/full
id doaj-527cb56792a14185abd7181c5d4e674b
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Xingyue Zhang
Xingyue Zhang
Lingfei Li
Lingfei Li
Qiong Zhang
Qiong Zhang
Qinglin Wei
Jiezhi Lin
Jiezhi Lin
Jiezhi Jia
Jiezhi Jia
Junhui Zhang
Junhui Zhang
Tiantian Yan
Yanling Lv
Yanling Lv
Xupin Jiang
Xupin Jiang
Peng Zhang
Peng Zhang
Huapei Song
Huapei Song
Dongxia Zhang
Dongxia Zhang
Yuesheng Huang
Yuesheng Huang
Yuesheng Huang
spellingShingle Xingyue Zhang
Xingyue Zhang
Lingfei Li
Lingfei Li
Qiong Zhang
Qiong Zhang
Qinglin Wei
Jiezhi Lin
Jiezhi Lin
Jiezhi Jia
Jiezhi Jia
Junhui Zhang
Junhui Zhang
Tiantian Yan
Yanling Lv
Yanling Lv
Xupin Jiang
Xupin Jiang
Peng Zhang
Peng Zhang
Huapei Song
Huapei Song
Dongxia Zhang
Dongxia Zhang
Yuesheng Huang
Yuesheng Huang
Yuesheng Huang
CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
Frontiers in Cell and Developmental Biology
pleckstrin homology domain-containing protein family member 1
Rab7
nicotinamide adenine dinucleotide
autophagosome–lysosome fusion
heart disease
author_facet Xingyue Zhang
Xingyue Zhang
Lingfei Li
Lingfei Li
Qiong Zhang
Qiong Zhang
Qinglin Wei
Jiezhi Lin
Jiezhi Lin
Jiezhi Jia
Jiezhi Jia
Junhui Zhang
Junhui Zhang
Tiantian Yan
Yanling Lv
Yanling Lv
Xupin Jiang
Xupin Jiang
Peng Zhang
Peng Zhang
Huapei Song
Huapei Song
Dongxia Zhang
Dongxia Zhang
Yuesheng Huang
Yuesheng Huang
Yuesheng Huang
author_sort Xingyue Zhang
title CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
title_short CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
title_full CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
title_fullStr CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
title_full_unstemmed CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia Conditions
title_sort cd38 causes autophagic flux inhibition and cardiac dysfunction through a transcriptional inhibition pathway under hypoxia/ischemia conditions
publisher Frontiers Media S.A.
series Frontiers in Cell and Developmental Biology
issn 2296-634X
publishDate 2020-04-01
description Induced autophagy is protective against myocardial hypoxia/ischemia (H/I) injury, but evidence regarding the extent of autophagic clearance under H/I and the molecular mechanisms that influence autophagic flux has scarcely been presented. Here, we report that CD38 knockout improved cardiac function and autophagic flux in CD38–/– mice and CD38–/– neonatal cardiomyocytes (CMs) under H/I conditions. Mechanistic studies demonstrated that overexpression of CD38 specifically downregulated the expression of Rab7 and its adaptor protein pleckstrin homology domain-containing protein family member 1 (PLEKHM1) through nicotinamide adenine dinucleotide (NAD)-dependent and non-NAD-dependent pathways, respectively. Loss of Rab7/PLEKHM1 impaired the fusion of autophagosomes and lysosomes, resulting in autophagosome accumulation in the myocardium and consequent cardiac dysfunction under H/I conditions. Thus, CD38 mediated autophagic flux blockade and cardiac dysfunction in a Rab7/PLEKHM1-dependent manner. These findings suggest a potential therapeutic strategy involving targeted suppression of CD38 expression.
topic pleckstrin homology domain-containing protein family member 1
Rab7
nicotinamide adenine dinucleotide
autophagosome–lysosome fusion
heart disease
url https://www.frontiersin.org/article/10.3389/fcell.2020.00191/full
work_keys_str_mv AT xingyuezhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT xingyuezhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT lingfeili cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT lingfeili cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT qiongzhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT qiongzhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT qinglinwei cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT jiezhilin cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT jiezhilin cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT jiezhijia cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT jiezhijia cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT junhuizhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT junhuizhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT tiantianyan cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT yanlinglv cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT yanlinglv cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT xupinjiang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT xupinjiang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT pengzhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT pengzhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT huapeisong cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT huapeisong cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT dongxiazhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT dongxiazhang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT yueshenghuang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT yueshenghuang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
AT yueshenghuang cd38causesautophagicfluxinhibitionandcardiacdysfunctionthroughatranscriptionalinhibitionpathwayunderhypoxiaischemiaconditions
_version_ 1724583615447171072
spelling doaj-527cb56792a14185abd7181c5d4e674b2020-11-25T03:28:31ZengFrontiers Media S.A.Frontiers in Cell and Developmental Biology2296-634X2020-04-01810.3389/fcell.2020.00191518830CD38 Causes Autophagic Flux Inhibition and Cardiac Dysfunction Through a Transcriptional Inhibition Pathway Under Hypoxia/Ischemia ConditionsXingyue Zhang0Xingyue Zhang1Lingfei Li2Lingfei Li3Qiong Zhang4Qiong Zhang5Qinglin Wei6Jiezhi Lin7Jiezhi Lin8Jiezhi Jia9Jiezhi Jia10Junhui Zhang11Junhui Zhang12Tiantian Yan13Yanling Lv14Yanling Lv15Xupin Jiang16Xupin Jiang17Peng Zhang18Peng Zhang19Huapei Song20Huapei Song21Dongxia Zhang22Dongxia Zhang23Yuesheng Huang24Yuesheng Huang25Yuesheng Huang26Institute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaCholestatic Liver Diseases Center of the Institute of Digestive Disease, First Affiliated of Army Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaMilitary Burn Center, The 990th (159th) Hospital of the People’s Liberation Army, Zhumadian, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaInstitute of Burn Research, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaState Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University, Third Military Medical University, Chongqing, ChinaDepartment of Wound Repair, Institute of Wound Repair, The First Affiliated Hospital of South University of Science and Technology (Shenzhen Peoples Hospital), Shenzhen, ChinaInduced autophagy is protective against myocardial hypoxia/ischemia (H/I) injury, but evidence regarding the extent of autophagic clearance under H/I and the molecular mechanisms that influence autophagic flux has scarcely been presented. Here, we report that CD38 knockout improved cardiac function and autophagic flux in CD38–/– mice and CD38–/– neonatal cardiomyocytes (CMs) under H/I conditions. Mechanistic studies demonstrated that overexpression of CD38 specifically downregulated the expression of Rab7 and its adaptor protein pleckstrin homology domain-containing protein family member 1 (PLEKHM1) through nicotinamide adenine dinucleotide (NAD)-dependent and non-NAD-dependent pathways, respectively. Loss of Rab7/PLEKHM1 impaired the fusion of autophagosomes and lysosomes, resulting in autophagosome accumulation in the myocardium and consequent cardiac dysfunction under H/I conditions. Thus, CD38 mediated autophagic flux blockade and cardiac dysfunction in a Rab7/PLEKHM1-dependent manner. These findings suggest a potential therapeutic strategy involving targeted suppression of CD38 expression.https://www.frontiersin.org/article/10.3389/fcell.2020.00191/fullpleckstrin homology domain-containing protein family member 1Rab7nicotinamide adenine dinucleotideautophagosome–lysosome fusionheart disease