Cargando…

Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis

We have previously reported that rhomboid domain containing 1 (RHBDD1), a mammalian rhomboid protease highly expressed in the testis, can cleave the Bcl-2 protein Bik. In this study, we identified a multi-pass transmembrane protein, tumor suppressor activated pathway-6 (TSAP6) as a potential substra...

Descripción completa

Detalles Bibliográficos
Autores principales: Wan, Chunhua, Fu, Jun, Wang, Yong, Miao, Shiying, Song, Wei, Wang, Linfang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3356283/
https://www.ncbi.nlm.nih.gov/pubmed/22624035
http://dx.doi.org/10.1371/journal.pone.0037452
_version_ 1782233536891191296
author Wan, Chunhua
Fu, Jun
Wang, Yong
Miao, Shiying
Song, Wei
Wang, Linfang
author_facet Wan, Chunhua
Fu, Jun
Wang, Yong
Miao, Shiying
Song, Wei
Wang, Linfang
author_sort Wan, Chunhua
collection PubMed
description We have previously reported that rhomboid domain containing 1 (RHBDD1), a mammalian rhomboid protease highly expressed in the testis, can cleave the Bcl-2 protein Bik. In this study, we identified a multi-pass transmembrane protein, tumor suppressor activated pathway-6 (TSAP6) as a potential substrate of RHBDD1. RHBDD1 was found to induce the proteolysis of TSAP6 in a dose- and activity-dependent manner. The cleavage of TSAP6 was not restricted to its glycosylated form and occurred in three different regions. In addition, mass spectrometry and mutagenesis analyses both indicated that the major cleavage site laid in the C-terminal of the third transmembrane domain of TSAP6. A somatic cell knock-in approach was used to genetically inactivate the endogenous RHBDD1 in HCT116 and RKO colon cancer cells. Exosome secretion was significantly elevated when RHBDD1 was inactivated in the two cells lines. The increased exosome secretion was verfied through the detection of certain exosomal components, including Tsg101, Tf-R, FasL and Trail. In addition, the elevation of exosome secretion by RHBDD1 inactivation was reduced when TSAP6 was knocked down, indicating that the role of RHBDD1 in regulating exosomal trafficking is very likely to be TSAP6-dependent. We found that the increase in FasL and Trail increased exosome-induced apoptosis in Jurkat cells. Taken together, our findings suggest that RHBDD1 is involved in the regulation of a nonclassical exosomal secretion pathway through the restriction of TSAP6.
format Online
Article
Text
id pubmed-3356283
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-33562832012-05-23 Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis Wan, Chunhua Fu, Jun Wang, Yong Miao, Shiying Song, Wei Wang, Linfang PLoS One Research Article We have previously reported that rhomboid domain containing 1 (RHBDD1), a mammalian rhomboid protease highly expressed in the testis, can cleave the Bcl-2 protein Bik. In this study, we identified a multi-pass transmembrane protein, tumor suppressor activated pathway-6 (TSAP6) as a potential substrate of RHBDD1. RHBDD1 was found to induce the proteolysis of TSAP6 in a dose- and activity-dependent manner. The cleavage of TSAP6 was not restricted to its glycosylated form and occurred in three different regions. In addition, mass spectrometry and mutagenesis analyses both indicated that the major cleavage site laid in the C-terminal of the third transmembrane domain of TSAP6. A somatic cell knock-in approach was used to genetically inactivate the endogenous RHBDD1 in HCT116 and RKO colon cancer cells. Exosome secretion was significantly elevated when RHBDD1 was inactivated in the two cells lines. The increased exosome secretion was verfied through the detection of certain exosomal components, including Tsg101, Tf-R, FasL and Trail. In addition, the elevation of exosome secretion by RHBDD1 inactivation was reduced when TSAP6 was knocked down, indicating that the role of RHBDD1 in regulating exosomal trafficking is very likely to be TSAP6-dependent. We found that the increase in FasL and Trail increased exosome-induced apoptosis in Jurkat cells. Taken together, our findings suggest that RHBDD1 is involved in the regulation of a nonclassical exosomal secretion pathway through the restriction of TSAP6. Public Library of Science 2012-05-18 /pmc/articles/PMC3356283/ /pubmed/22624035 http://dx.doi.org/10.1371/journal.pone.0037452 Text en Wan et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Wan, Chunhua
Fu, Jun
Wang, Yong
Miao, Shiying
Song, Wei
Wang, Linfang
Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title_full Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title_fullStr Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title_full_unstemmed Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title_short Exosome-Related Multi-Pass Transmembrane Protein TSAP6 Is a Target of Rhomboid Protease RHBDD1-Induced Proteolysis
title_sort exosome-related multi-pass transmembrane protein tsap6 is a target of rhomboid protease rhbdd1-induced proteolysis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3356283/
https://www.ncbi.nlm.nih.gov/pubmed/22624035
http://dx.doi.org/10.1371/journal.pone.0037452
work_keys_str_mv AT wanchunhua exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis
AT fujun exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis
AT wangyong exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis
AT miaoshiying exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis
AT songwei exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis
AT wanglinfang exosomerelatedmultipasstransmembraneproteintsap6isatargetofrhomboidproteaserhbdd1inducedproteolysis