Cargando…

Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells

The mucin 1 (MUC1) gene emerged in mammals to afford protection of barrier epithelial tissues from the external environment. MUC1 encodes a transmembrane C-terminal (MUC1-C) subunit that is activated by loss of homeostasis and induces inflammatory, proliferative, and remodeling pathways associated w...

Descripción completa

Detalles Bibliográficos
Autor principal: Kufe, Donald W.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9083497/
https://www.ncbi.nlm.nih.gov/pubmed/35539431
http://dx.doi.org/10.20517/2394-4722.2022.03
_version_ 1784703434095591424
author Kufe, Donald W.
author_facet Kufe, Donald W.
author_sort Kufe, Donald W.
collection PubMed
description The mucin 1 (MUC1) gene emerged in mammals to afford protection of barrier epithelial tissues from the external environment. MUC1 encodes a transmembrane C-terminal (MUC1-C) subunit that is activated by loss of homeostasis and induces inflammatory, proliferative, and remodeling pathways associated with wound repair. As a consequence, chronic activation of MUC1-C promotes lineage plasticity, epigenetic reprogramming, and carcinogenesis. In driving cancer progression, MUC1-C is imported into the nucleus, where it induces NF-κB inflammatory signaling and the epithelial-mesenchymal transition (EMT). MUC1-C represses gene expression by activating (i) DNA methyltransferase 1 (DNMT1) and DNMT3b, (ii) Polycomb Repressive Complex 1 (PRC1) and PRC2, and (iii) the nucleosome remodeling and deacetylase (NuRD) complex. PRC1/2-mediated gene repression is counteracted by the SWI/SNF chromatin remodeling complexes. MUC1-C activates the SWI/SNF BAF and PBAF complexes in cancer stem cell (CSC) models with the induction of genome-wide differentially accessible regions and expressed genes. MUC1-C regulates chromatin accessibility of enhancer-like signatures in association with the induction of the Yamanaka pluripotency factors and recruitment of JUN and BAF, which promote increases in histone activation marks and opening of chromatin. These and other findings described in this review have uncovered a pivotal role for MUC1-C in integrating lineage plasticity and epigenetic reprogramming, which are transient in wound repair and sustained in promoting CSC progression.
format Online
Article
Text
id pubmed-9083497
institution National Center for Biotechnology Information
language English
publishDate 2022
record_format MEDLINE/PubMed
spelling pubmed-90834972022-05-09 Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells Kufe, Donald W. J Cancer Metastasis Treat Article The mucin 1 (MUC1) gene emerged in mammals to afford protection of barrier epithelial tissues from the external environment. MUC1 encodes a transmembrane C-terminal (MUC1-C) subunit that is activated by loss of homeostasis and induces inflammatory, proliferative, and remodeling pathways associated with wound repair. As a consequence, chronic activation of MUC1-C promotes lineage plasticity, epigenetic reprogramming, and carcinogenesis. In driving cancer progression, MUC1-C is imported into the nucleus, where it induces NF-κB inflammatory signaling and the epithelial-mesenchymal transition (EMT). MUC1-C represses gene expression by activating (i) DNA methyltransferase 1 (DNMT1) and DNMT3b, (ii) Polycomb Repressive Complex 1 (PRC1) and PRC2, and (iii) the nucleosome remodeling and deacetylase (NuRD) complex. PRC1/2-mediated gene repression is counteracted by the SWI/SNF chromatin remodeling complexes. MUC1-C activates the SWI/SNF BAF and PBAF complexes in cancer stem cell (CSC) models with the induction of genome-wide differentially accessible regions and expressed genes. MUC1-C regulates chromatin accessibility of enhancer-like signatures in association with the induction of the Yamanaka pluripotency factors and recruitment of JUN and BAF, which promote increases in histone activation marks and opening of chromatin. These and other findings described in this review have uncovered a pivotal role for MUC1-C in integrating lineage plasticity and epigenetic reprogramming, which are transient in wound repair and sustained in promoting CSC progression. 2022 2022-03-31 /pmc/articles/PMC9083497/ /pubmed/35539431 http://dx.doi.org/10.20517/2394-4722.2022.03 Text en https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
spellingShingle Article
Kufe, Donald W.
Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title_full Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title_fullStr Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title_full_unstemmed Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title_short Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells
title_sort chronic activation of muc1-c in wound repair promotes progression to cancer stem cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9083497/
https://www.ncbi.nlm.nih.gov/pubmed/35539431
http://dx.doi.org/10.20517/2394-4722.2022.03
work_keys_str_mv AT kufedonaldw chronicactivationofmuc1cinwoundrepairpromotesprogressiontocancerstemcells