Cargando…

Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals

Mucins are functionally implicated in a range of human pathologies, including cystic fibrosis, influenza, bacterial endocarditis, gut dysbiosis, and cancer. These observations have motivated the study of mucin biosynthesis as well as the development of strategies for inhibition of mucin glycosylatio...

Descripción completa

Detalles Bibliográficos
Autores principales: Pedram, Kayvon, Laqtom, Nouf N., Shon, D. Judy, Di Spiezio, Alessandro, Riley, Nicholas M., Saftig, Paul, Abu-Remaileh, Monther, Bertozzi, Carolyn R.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: National Academy of Sciences 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9522329/
https://www.ncbi.nlm.nih.gov/pubmed/36122205
http://dx.doi.org/10.1073/pnas.2117105119
_version_ 1784800040915566592
author Pedram, Kayvon
Laqtom, Nouf N.
Shon, D. Judy
Di Spiezio, Alessandro
Riley, Nicholas M.
Saftig, Paul
Abu-Remaileh, Monther
Bertozzi, Carolyn R.
author_facet Pedram, Kayvon
Laqtom, Nouf N.
Shon, D. Judy
Di Spiezio, Alessandro
Riley, Nicholas M.
Saftig, Paul
Abu-Remaileh, Monther
Bertozzi, Carolyn R.
author_sort Pedram, Kayvon
collection PubMed
description Mucins are functionally implicated in a range of human pathologies, including cystic fibrosis, influenza, bacterial endocarditis, gut dysbiosis, and cancer. These observations have motivated the study of mucin biosynthesis as well as the development of strategies for inhibition of mucin glycosylation. Mammalian pathways for mucin catabolism, however, have remained underexplored. The canonical view, derived from analysis of N-glycoproteins in human lysosomal storage disorders, is that glycan degradation and proteolysis occur sequentially. Here, we challenge this view by providing genetic and biochemical evidence supporting mammalian proteolysis of heavily O-glycosylated mucin domains without prior deglycosylation. Using activity screening coupled with mass spectrometry, we ascribed mucin-degrading activity in murine liver to the lysosomal protease cathepsin D. Glycoproteomics of substrates digested with purified human liver lysosomal cathepsin D provided direct evidence for proteolysis within densely O-glycosylated domains. Finally, knockout of cathepsin D in a murine model of the human lysosomal storage disorder neuronal ceroid lipofuscinosis 10 resulted in accumulation of mucins in liver-resident macrophages. Our findings imply that mucin-degrading activity is a component of endogenous pathways for glycoprotein catabolism in mammalian tissues.
format Online
Article
Text
id pubmed-9522329
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher National Academy of Sciences
record_format MEDLINE/PubMed
spelling pubmed-95223292022-09-30 Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals Pedram, Kayvon Laqtom, Nouf N. Shon, D. Judy Di Spiezio, Alessandro Riley, Nicholas M. Saftig, Paul Abu-Remaileh, Monther Bertozzi, Carolyn R. Proc Natl Acad Sci U S A Biological Sciences Mucins are functionally implicated in a range of human pathologies, including cystic fibrosis, influenza, bacterial endocarditis, gut dysbiosis, and cancer. These observations have motivated the study of mucin biosynthesis as well as the development of strategies for inhibition of mucin glycosylation. Mammalian pathways for mucin catabolism, however, have remained underexplored. The canonical view, derived from analysis of N-glycoproteins in human lysosomal storage disorders, is that glycan degradation and proteolysis occur sequentially. Here, we challenge this view by providing genetic and biochemical evidence supporting mammalian proteolysis of heavily O-glycosylated mucin domains without prior deglycosylation. Using activity screening coupled with mass spectrometry, we ascribed mucin-degrading activity in murine liver to the lysosomal protease cathepsin D. Glycoproteomics of substrates digested with purified human liver lysosomal cathepsin D provided direct evidence for proteolysis within densely O-glycosylated domains. Finally, knockout of cathepsin D in a murine model of the human lysosomal storage disorder neuronal ceroid lipofuscinosis 10 resulted in accumulation of mucins in liver-resident macrophages. Our findings imply that mucin-degrading activity is a component of endogenous pathways for glycoprotein catabolism in mammalian tissues. National Academy of Sciences 2022-09-19 2022-09-27 /pmc/articles/PMC9522329/ /pubmed/36122205 http://dx.doi.org/10.1073/pnas.2117105119 Text en Copyright © 2022 the Author(s). Published by PNAS. https://creativecommons.org/licenses/by-nc-nd/4.0/This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND) (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Biological Sciences
Pedram, Kayvon
Laqtom, Nouf N.
Shon, D. Judy
Di Spiezio, Alessandro
Riley, Nicholas M.
Saftig, Paul
Abu-Remaileh, Monther
Bertozzi, Carolyn R.
Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title_full Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title_fullStr Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title_full_unstemmed Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title_short Lysosomal cathepsin D mediates endogenous mucin glycodomain catabolism in mammals
title_sort lysosomal cathepsin d mediates endogenous mucin glycodomain catabolism in mammals
topic Biological Sciences
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9522329/
https://www.ncbi.nlm.nih.gov/pubmed/36122205
http://dx.doi.org/10.1073/pnas.2117105119
work_keys_str_mv AT pedramkayvon lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT laqtomnoufn lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT shondjudy lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT dispiezioalessandro lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT rileynicholasm lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT saftigpaul lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT aburemailehmonther lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals
AT bertozzicarolynr lysosomalcathepsindmediatesendogenousmucinglycodomaincatabolisminmammals