Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction

Abstract Background The autophagy adapter SQSTM1/p62 is crucial for maintaining homeostasis in various organs and cells due to its protein–protein interaction domains and involvement in diverse physiological and pathological processes. Vascular endothelium cells play a unique role in vascular biolog...

Full description

Bibliographic Details
Main Authors: Jing Feng, Yan Li, Yu Zhang, Shengnan Sun, Jian Sun, Quanlin Xu, Xingzhao Ji, Yi Liu, Qiang Wan
Format: Article
Language:English
Published: BMC 2024-02-01
Series:Journal of Translational Medicine
Subjects:
Online Access:https://doi.org/10.1186/s12967-024-04946-w
_version_ 1797273500603383808
author Jing Feng
Yan Li
Yu Zhang
Shengnan Sun
Jian Sun
Quanlin Xu
Xingzhao Ji
Yi Liu
Qiang Wan
author_facet Jing Feng
Yan Li
Yu Zhang
Shengnan Sun
Jian Sun
Quanlin Xu
Xingzhao Ji
Yi Liu
Qiang Wan
author_sort Jing Feng
collection DOAJ
description Abstract Background The autophagy adapter SQSTM1/p62 is crucial for maintaining homeostasis in various organs and cells due to its protein–protein interaction domains and involvement in diverse physiological and pathological processes. Vascular endothelium cells play a unique role in vascular biology and contribute to vascular health. Methods Using the Cre-loxP system, we generated mice with endothelium cell-specific knockout of p62 mediated by Tek (Tek receptor tyrosine kinase)-cre to investigate the essential role of p62 in the endothelium. In vitro, we employed protein mass spectrometry and IPA to identify differentially expressed proteins upon knockdown of p62. Immunoprecipitation assays were conducted to demonstrate the interaction between p62 and FN1 or LAMC2 in human umbilical vein endothelium cells (HUVECs). Additionally, we identified the degradation pathway of FN1 and LAMC2 using the autophagy inhibitor 3-methyladenine (3-MA) or proteasome inhibitor MG132. Finally, the results of immunoprecipitation demonstrated that the interaction between p62 and LAMC2 was abolished in the PB1 truncation group of p62, while the interaction between p62 and FN1 was abolished in the UBA truncation group of p62. Results Our findings revealed that p62 Endo mice exhibited heart, lung, and kidney fibrosis compared to littermate controls, accompanied by severe cardiac dysfunction. Immunoprecipitation assays provided evidence of p62 acting as an autophagy adapter in the autophagy-lysosome pathway for FN1 and LAMC2 degradation respectively through PB1 and UBA domain with these proteins rather than proteasome system. Conclusions Our study demonstrates that defects in p62 within endothelium cells induce multi-organ fibrosis and cardiac dysfunction in mice. Our findings indicate that FN1 and LAMC2, as markers of (EndoMT), have detrimental effects on HUVECs and elucidate the autophagy-lysosome degradation mechanism of FN1 and LAMC2.
first_indexed 2024-03-07T14:44:12Z
format Article
id doaj.art-f6181a889fb940d58565ec1a52d533a4
institution Directory Open Access Journal
issn 1479-5876
language English
last_indexed 2024-03-07T14:44:12Z
publishDate 2024-02-01
publisher BMC
record_format Article
series Journal of Translational Medicine
spelling doaj.art-f6181a889fb940d58565ec1a52d533a42024-03-05T20:06:37ZengBMCJournal of Translational Medicine1479-58762024-02-0122111810.1186/s12967-024-04946-wEndothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunctionJing Feng0Yan Li1Yu Zhang2Shengnan Sun3Jian Sun4Quanlin Xu5Xingzhao Ji6Yi Liu7Qiang Wan8Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong UniversityDepartment of Pulmonary and Critical Care Medicine, The Second Hospital of Shandong UniversityDepartment of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Shandong UniversityKey Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong UniversityDepartment of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityDepartment of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityDepartment of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityDepartment of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Shandong UniversityKey Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Jinan Central Hospital, Shandong UniversityAbstract Background The autophagy adapter SQSTM1/p62 is crucial for maintaining homeostasis in various organs and cells due to its protein–protein interaction domains and involvement in diverse physiological and pathological processes. Vascular endothelium cells play a unique role in vascular biology and contribute to vascular health. Methods Using the Cre-loxP system, we generated mice with endothelium cell-specific knockout of p62 mediated by Tek (Tek receptor tyrosine kinase)-cre to investigate the essential role of p62 in the endothelium. In vitro, we employed protein mass spectrometry and IPA to identify differentially expressed proteins upon knockdown of p62. Immunoprecipitation assays were conducted to demonstrate the interaction between p62 and FN1 or LAMC2 in human umbilical vein endothelium cells (HUVECs). Additionally, we identified the degradation pathway of FN1 and LAMC2 using the autophagy inhibitor 3-methyladenine (3-MA) or proteasome inhibitor MG132. Finally, the results of immunoprecipitation demonstrated that the interaction between p62 and LAMC2 was abolished in the PB1 truncation group of p62, while the interaction between p62 and FN1 was abolished in the UBA truncation group of p62. Results Our findings revealed that p62 Endo mice exhibited heart, lung, and kidney fibrosis compared to littermate controls, accompanied by severe cardiac dysfunction. Immunoprecipitation assays provided evidence of p62 acting as an autophagy adapter in the autophagy-lysosome pathway for FN1 and LAMC2 degradation respectively through PB1 and UBA domain with these proteins rather than proteasome system. Conclusions Our study demonstrates that defects in p62 within endothelium cells induce multi-organ fibrosis and cardiac dysfunction in mice. Our findings indicate that FN1 and LAMC2, as markers of (EndoMT), have detrimental effects on HUVECs and elucidate the autophagy-lysosome degradation mechanism of FN1 and LAMC2.https://doi.org/10.1186/s12967-024-04946-wSQSTM1/p62Endothelium cellsFibrosisCardiac dysfunctionFibronectin1Lamininγ2
spellingShingle Jing Feng
Yan Li
Yu Zhang
Shengnan Sun
Jian Sun
Quanlin Xu
Xingzhao Ji
Yi Liu
Qiang Wan
Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
Journal of Translational Medicine
SQSTM1/p62
Endothelium cells
Fibrosis
Cardiac dysfunction
Fibronectin1
Lamininγ2
title Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
title_full Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
title_fullStr Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
title_full_unstemmed Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
title_short Endothelium-specific deletion of p62 causes organ fibrosis and cardiac dysfunction
title_sort endothelium specific deletion of p62 causes organ fibrosis and cardiac dysfunction
topic SQSTM1/p62
Endothelium cells
Fibrosis
Cardiac dysfunction
Fibronectin1
Lamininγ2
url https://doi.org/10.1186/s12967-024-04946-w
work_keys_str_mv AT jingfeng endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT yanli endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT yuzhang endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT shengnansun endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT jiansun endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT quanlinxu endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT xingzhaoji endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT yiliu endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction
AT qiangwan endotheliumspecificdeletionofp62causesorganfibrosisandcardiacdysfunction