Cargando…

NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival

DNA damage signaling is critical for the maintenance of genome integrity and cell fate decision. Poly(ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor rapidly activated in a damage dose- and complexity-dependent manner playing a critical role in the initial chromatin organization and DNA repa...

Descripción completa

Detalles Bibliográficos
Autores principales: Murata, Michael M., Kong, Xiangduo, Moncada, Emmanuel, Chen, Yumay, Imamura, Hiromi, Wang, Ping, Berns, Michael W., Yokomori, Kyoko, Digman, Michelle A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: The American Society for Cell Biology 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6740200/
https://www.ncbi.nlm.nih.gov/pubmed/31390283
http://dx.doi.org/10.1091/mbc.E18-10-0650
_version_ 1783451056275980288
author Murata, Michael M.
Kong, Xiangduo
Moncada, Emmanuel
Chen, Yumay
Imamura, Hiromi
Wang, Ping
Berns, Michael W.
Yokomori, Kyoko
Digman, Michelle A.
author_facet Murata, Michael M.
Kong, Xiangduo
Moncada, Emmanuel
Chen, Yumay
Imamura, Hiromi
Wang, Ping
Berns, Michael W.
Yokomori, Kyoko
Digman, Michelle A.
author_sort Murata, Michael M.
collection PubMed
description DNA damage signaling is critical for the maintenance of genome integrity and cell fate decision. Poly(ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor rapidly activated in a damage dose- and complexity-dependent manner playing a critical role in the initial chromatin organization and DNA repair pathway choice at damage sites. However, our understanding of a cell-wide consequence of its activation in damaged cells is still limited. Using the phasor approach to fluorescence lifetime imaging microscopy and fluorescence-based biosensors in combination with laser microirradiation, we found a rapid cell-wide increase of the bound NADH fraction in response to nuclear DNA damage, which is triggered by PARP-dependent NAD+ depletion. This change is linked to the metabolic balance shift to oxidative phosphorylation (oxphos) over glycolysis. Inhibition of oxphos, but not glycolysis, resulted in parthanatos due to rapid PARP-dependent ATP deprivation, indicating that oxphos becomes critical for damaged cell survival. The results reveal the novel prosurvival response to PARP activation through a change in cellular metabolism and demonstrate how unique applications of advanced fluorescence imaging and laser microirradiation-induced DNA damage can be a powerful tool to interrogate damage-induced metabolic changes at high spatiotemporal resolution in a live cell.
format Online
Article
Text
id pubmed-6740200
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher The American Society for Cell Biology
record_format MEDLINE/PubMed
spelling pubmed-67402002019-11-30 NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival Murata, Michael M. Kong, Xiangduo Moncada, Emmanuel Chen, Yumay Imamura, Hiromi Wang, Ping Berns, Michael W. Yokomori, Kyoko Digman, Michelle A. Mol Biol Cell Articles DNA damage signaling is critical for the maintenance of genome integrity and cell fate decision. Poly(ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor rapidly activated in a damage dose- and complexity-dependent manner playing a critical role in the initial chromatin organization and DNA repair pathway choice at damage sites. However, our understanding of a cell-wide consequence of its activation in damaged cells is still limited. Using the phasor approach to fluorescence lifetime imaging microscopy and fluorescence-based biosensors in combination with laser microirradiation, we found a rapid cell-wide increase of the bound NADH fraction in response to nuclear DNA damage, which is triggered by PARP-dependent NAD+ depletion. This change is linked to the metabolic balance shift to oxidative phosphorylation (oxphos) over glycolysis. Inhibition of oxphos, but not glycolysis, resulted in parthanatos due to rapid PARP-dependent ATP deprivation, indicating that oxphos becomes critical for damaged cell survival. The results reveal the novel prosurvival response to PARP activation through a change in cellular metabolism and demonstrate how unique applications of advanced fluorescence imaging and laser microirradiation-induced DNA damage can be a powerful tool to interrogate damage-induced metabolic changes at high spatiotemporal resolution in a live cell. The American Society for Cell Biology 2019-09-15 /pmc/articles/PMC6740200/ /pubmed/31390283 http://dx.doi.org/10.1091/mbc.E18-10-0650 Text en © 2019 Murata, Kong, et al. “ASCB®,” “The American Society for Cell Biology®,” and “Molecular Biology of the Cell®” are registered trademarks of The American Society for Cell Biology. http://creativecommons.org/licenses/by-nc-sa/3.0 This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License.
spellingShingle Articles
Murata, Michael M.
Kong, Xiangduo
Moncada, Emmanuel
Chen, Yumay
Imamura, Hiromi
Wang, Ping
Berns, Michael W.
Yokomori, Kyoko
Digman, Michelle A.
NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title_full NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title_fullStr NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title_full_unstemmed NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title_short NAD+ consumption by PARP1 in response to DNA damage triggers metabolic shift critical for damaged cell survival
title_sort nad+ consumption by parp1 in response to dna damage triggers metabolic shift critical for damaged cell survival
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6740200/
https://www.ncbi.nlm.nih.gov/pubmed/31390283
http://dx.doi.org/10.1091/mbc.E18-10-0650
work_keys_str_mv AT muratamichaelm nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT kongxiangduo nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT moncadaemmanuel nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT chenyumay nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT imamurahiromi nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT wangping nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT bernsmichaelw nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT yokomorikyoko nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival
AT digmanmichellea nadconsumptionbyparp1inresponsetodnadamagetriggersmetabolicshiftcriticalfordamagedcellsurvival