Cargando…

Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging

BACKGROUND: Accumulation of DNA damage leading to adult stem cell exhaustion has been proposed to be a principal mechanism of aging. Here we tested this hypothesis in healthy individuals of different ages by examining unrepaired DNA double-strand breaks (DSBs) in hematopoietic stem/progenitor cells...

Descripción completa

Detalles Bibliográficos
Autores principales: Rübe, Claudia E., Fricke, Andreas, Widmann, Thomas A., Fürst, Tobias, Madry, Henning, Pfreundschuh, Michael, Rübe, Christian
Formato: Texto
Lenguaje:English
Publicado: Public Library of Science 2011
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3049780/
https://www.ncbi.nlm.nih.gov/pubmed/21408175
http://dx.doi.org/10.1371/journal.pone.0017487
_version_ 1782199273228599296
author Rübe, Claudia E.
Fricke, Andreas
Widmann, Thomas A.
Fürst, Tobias
Madry, Henning
Pfreundschuh, Michael
Rübe, Christian
author_facet Rübe, Claudia E.
Fricke, Andreas
Widmann, Thomas A.
Fürst, Tobias
Madry, Henning
Pfreundschuh, Michael
Rübe, Christian
author_sort Rübe, Claudia E.
collection PubMed
description BACKGROUND: Accumulation of DNA damage leading to adult stem cell exhaustion has been proposed to be a principal mechanism of aging. Here we tested this hypothesis in healthy individuals of different ages by examining unrepaired DNA double-strand breaks (DSBs) in hematopoietic stem/progenitor cells matured in their physiological microenvironment. METHODOLOGY/PRINCIPAL FINDINGS: To asses DNA damage accumulation and repair capacities, γH2AX-foci were examined before and after exposure to ionizing irradiation. Analyzing CD34+ and CD34− stem/progenitor cells we observed an increase of endogenous γH2AX-foci levels with advancing donor age, associated with an age-related decline in telomere length. Using combined immunofluorescence and telomere-fluorescence in-situ hybridization we show that γH2AX-foci co-localize consistently with other repair factors such as pATM, MDC1 and 53BP1, but not significantly with telomeres, strongly supporting the telomere-independent origin for the majority of foci. The highest inter-individual variations for non-telomeric DNA damage were observed in middle-aged donors, whereas the individual DSB repair capacity appears to determine the extent of DNA damage accrual. However, analyzing different stem/progenitor subpopulations obtained from healthy elderly (>70 years), we observed an only modest increase in DNA damage accrual, most pronounced in the primitive CD34+CD38−-enriched subfraction, but sustained DNA repair efficiencies, suggesting that healthy lifestyle may slow down the natural aging process. CONCLUSIONS/SIGNIFICANCE: Based on these findings we conclude that age-related non-telomeric DNA damage accrual accompanies physiological stem cell aging in humans. Moreover, aging may alter the functional capacity of human stem cells to repair DSBs, thereby deteriorating an important genome protection mechanism leading to exceeding DNA damage accumulation. However, the great inter-individual variations in middle-aged individuals suggest that additional cell-intrinsic mechanisms and/or extrinsic factors contribute to the age-associated DNA damage accumulation.
format Text
id pubmed-3049780
institution National Center for Biotechnology Information
language English
publishDate 2011
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-30497802011-03-15 Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging Rübe, Claudia E. Fricke, Andreas Widmann, Thomas A. Fürst, Tobias Madry, Henning Pfreundschuh, Michael Rübe, Christian PLoS One Research Article BACKGROUND: Accumulation of DNA damage leading to adult stem cell exhaustion has been proposed to be a principal mechanism of aging. Here we tested this hypothesis in healthy individuals of different ages by examining unrepaired DNA double-strand breaks (DSBs) in hematopoietic stem/progenitor cells matured in their physiological microenvironment. METHODOLOGY/PRINCIPAL FINDINGS: To asses DNA damage accumulation and repair capacities, γH2AX-foci were examined before and after exposure to ionizing irradiation. Analyzing CD34+ and CD34− stem/progenitor cells we observed an increase of endogenous γH2AX-foci levels with advancing donor age, associated with an age-related decline in telomere length. Using combined immunofluorescence and telomere-fluorescence in-situ hybridization we show that γH2AX-foci co-localize consistently with other repair factors such as pATM, MDC1 and 53BP1, but not significantly with telomeres, strongly supporting the telomere-independent origin for the majority of foci. The highest inter-individual variations for non-telomeric DNA damage were observed in middle-aged donors, whereas the individual DSB repair capacity appears to determine the extent of DNA damage accrual. However, analyzing different stem/progenitor subpopulations obtained from healthy elderly (>70 years), we observed an only modest increase in DNA damage accrual, most pronounced in the primitive CD34+CD38−-enriched subfraction, but sustained DNA repair efficiencies, suggesting that healthy lifestyle may slow down the natural aging process. CONCLUSIONS/SIGNIFICANCE: Based on these findings we conclude that age-related non-telomeric DNA damage accrual accompanies physiological stem cell aging in humans. Moreover, aging may alter the functional capacity of human stem cells to repair DSBs, thereby deteriorating an important genome protection mechanism leading to exceeding DNA damage accumulation. However, the great inter-individual variations in middle-aged individuals suggest that additional cell-intrinsic mechanisms and/or extrinsic factors contribute to the age-associated DNA damage accumulation. Public Library of Science 2011-03-07 /pmc/articles/PMC3049780/ /pubmed/21408175 http://dx.doi.org/10.1371/journal.pone.0017487 Text en This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration which stipulates that, once placed in the public domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. https://creativecommons.org/publicdomain/zero/1.0/ This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration, which stipulates that, once placed in the public domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose.
spellingShingle Research Article
Rübe, Claudia E.
Fricke, Andreas
Widmann, Thomas A.
Fürst, Tobias
Madry, Henning
Pfreundschuh, Michael
Rübe, Christian
Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title_full Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title_fullStr Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title_full_unstemmed Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title_short Accumulation of DNA Damage in Hematopoietic Stem and Progenitor Cells during Human Aging
title_sort accumulation of dna damage in hematopoietic stem and progenitor cells during human aging
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3049780/
https://www.ncbi.nlm.nih.gov/pubmed/21408175
http://dx.doi.org/10.1371/journal.pone.0017487
work_keys_str_mv AT rubeclaudiae accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT frickeandreas accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT widmannthomasa accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT fursttobias accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT madryhenning accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT pfreundschuhmichael accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging
AT rubechristian accumulationofdnadamageinhematopoieticstemandprogenitorcellsduringhumanaging