Cargando…

p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging

Hematopoietic stem cells (HSCs) are characterized by the capacity for self-renewal and the ability to reconstitute the entire hematopoietic compartment. Thrombopoietin maintains adult HSCs in a quiescent state through the induction of cell cycle inhibitors p57(Kip2) and p19(INK4d). Using the p19(INK...

Descripción completa

Detalles Bibliográficos
Autores principales: Hilpert, Morgane, Legrand, Céline, Bluteau, Dominique, Balayn, Natalie, Betems, Aline, Bluteau, Olivier, Villeval, Jean-Luc, Louache, Fawzia, Gonin, Patrick, Debili, Najet, Plo, Isabelle, Vainchenker, William, Gilles, Laure, Raslova, Hana
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4264042/
https://www.ncbi.nlm.nih.gov/pubmed/25458892
http://dx.doi.org/10.1016/j.stemcr.2014.10.005
_version_ 1782348658782502912
author Hilpert, Morgane
Legrand, Céline
Bluteau, Dominique
Balayn, Natalie
Betems, Aline
Bluteau, Olivier
Villeval, Jean-Luc
Louache, Fawzia
Gonin, Patrick
Debili, Najet
Plo, Isabelle
Vainchenker, William
Gilles, Laure
Raslova, Hana
author_facet Hilpert, Morgane
Legrand, Céline
Bluteau, Dominique
Balayn, Natalie
Betems, Aline
Bluteau, Olivier
Villeval, Jean-Luc
Louache, Fawzia
Gonin, Patrick
Debili, Najet
Plo, Isabelle
Vainchenker, William
Gilles, Laure
Raslova, Hana
author_sort Hilpert, Morgane
collection PubMed
description Hematopoietic stem cells (HSCs) are characterized by the capacity for self-renewal and the ability to reconstitute the entire hematopoietic compartment. Thrombopoietin maintains adult HSCs in a quiescent state through the induction of cell cycle inhibitors p57(Kip2) and p19(INK4d). Using the p19(INK4d−/−) mouse model, we investigated the role of p19(INK4d) in basal and stress-induced hematopoiesis. We demonstrate that p19(INK4d) is involved in the regulation of HSC quiescence by inhibition of the G0/G1 cell cycle transition. Under genotoxic stress conditions, the absence of p19(INK4d) in HSCs leads to accelerated cell cycle exit, accumulation of DNA double-strand breaks, and apoptosis when cells progress to the S/G2-M stages of the cell cycle. Moreover, p19(INK4d) controls the HSC microenvironment through negative regulation of megakaryopoiesis. Deletion of p19(INK4d) results in megakaryocyte hyperproliferation and increased transforming growth factor β1 secretion. This leads to fibrosis in the bone marrow and spleen, followed by loss of HSCs during aging.
format Online
Article
Text
id pubmed-4264042
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-42640422014-12-13 p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging Hilpert, Morgane Legrand, Céline Bluteau, Dominique Balayn, Natalie Betems, Aline Bluteau, Olivier Villeval, Jean-Luc Louache, Fawzia Gonin, Patrick Debili, Najet Plo, Isabelle Vainchenker, William Gilles, Laure Raslova, Hana Stem Cell Reports Article Hematopoietic stem cells (HSCs) are characterized by the capacity for self-renewal and the ability to reconstitute the entire hematopoietic compartment. Thrombopoietin maintains adult HSCs in a quiescent state through the induction of cell cycle inhibitors p57(Kip2) and p19(INK4d). Using the p19(INK4d−/−) mouse model, we investigated the role of p19(INK4d) in basal and stress-induced hematopoiesis. We demonstrate that p19(INK4d) is involved in the regulation of HSC quiescence by inhibition of the G0/G1 cell cycle transition. Under genotoxic stress conditions, the absence of p19(INK4d) in HSCs leads to accelerated cell cycle exit, accumulation of DNA double-strand breaks, and apoptosis when cells progress to the S/G2-M stages of the cell cycle. Moreover, p19(INK4d) controls the HSC microenvironment through negative regulation of megakaryopoiesis. Deletion of p19(INK4d) results in megakaryocyte hyperproliferation and increased transforming growth factor β1 secretion. This leads to fibrosis in the bone marrow and spleen, followed by loss of HSCs during aging. Elsevier 2014-11-20 /pmc/articles/PMC4264042/ /pubmed/25458892 http://dx.doi.org/10.1016/j.stemcr.2014.10.005 Text en © 2014 The Authors http://creativecommons.org/licenses/by-nc-nd/3.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
spellingShingle Article
Hilpert, Morgane
Legrand, Céline
Bluteau, Dominique
Balayn, Natalie
Betems, Aline
Bluteau, Olivier
Villeval, Jean-Luc
Louache, Fawzia
Gonin, Patrick
Debili, Najet
Plo, Isabelle
Vainchenker, William
Gilles, Laure
Raslova, Hana
p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title_full p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title_fullStr p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title_full_unstemmed p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title_short p19(INK4d) Controls Hematopoietic Stem Cells in a Cell-Autonomous Manner during Genotoxic Stress and through the Microenvironment during Aging
title_sort p19(ink4d) controls hematopoietic stem cells in a cell-autonomous manner during genotoxic stress and through the microenvironment during aging
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4264042/
https://www.ncbi.nlm.nih.gov/pubmed/25458892
http://dx.doi.org/10.1016/j.stemcr.2014.10.005
work_keys_str_mv AT hilpertmorgane p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT legrandceline p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT bluteaudominique p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT balaynnatalie p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT betemsaline p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT bluteauolivier p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT villevaljeanluc p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT louachefawzia p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT goninpatrick p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT debilinajet p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT ploisabelle p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT vainchenkerwilliam p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT gilleslaure p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging
AT raslovahana p19ink4dcontrolshematopoieticstemcellsinacellautonomousmannerduringgenotoxicstressandthroughthemicroenvironmentduringaging