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

J Transl Med. 2024 Feb 16;22(1):161. doi: 10.1186/s12967-024-04946-w.

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.

Keywords: Cardiac dysfunction; Endothelium cells; Fibronectin1; Fibrosis; Lamininγ2; SQSTM1/p62.

MeSH terms

  • Adaptor Proteins, Signal Transducing / metabolism
  • Animals
  • Autophagy
  • Endothelium / metabolism
  • Fibrosis / genetics
  • Fibrosis / metabolism
  • Heart Diseases* / genetics
  • Heart Diseases* / metabolism
  • Humans
  • Mice
  • Proteasome Endopeptidase Complex / metabolism
  • Proteasome Endopeptidase Complex / pharmacology
  • Sequestosome-1 Protein* / genetics
  • Sequestosome-1 Protein* / metabolism

Substances

  • Adaptor Proteins, Signal Transducing
  • Proteasome Endopeptidase Complex
  • Sequestosome-1 Protein