Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Diminished apoptotic priming and ATM signalling confer a survival advantage onto aged haematopoietic stem cells in response to DNA damage

Abstract

Ageing of haematopoietic stem cells (HSCs) contributes to deficits in the aged haematopoietic system. HSC decline is driven in part by DNA damage accumulation; yet, how ageing impacts the acute DNA damage response (DDR) of HSCs is poorly understood. We show that old HSCs exhibit diminished ATM activity and attenuated DDR, leading to elevated clonal survival in response to a range of genotoxins that was underwritten by diminished apoptotic priming. Distinct HSC subsets exhibited ageing-dependent and subtype-dependent differences in apoptotic priming and survival in response to DNA damage. The defective DDR of old HSCs was non-cell autonomous, as ATM signalling and clonal survival in response to DNA damage could be restored to levels observed in young HSCs post-transplantated into young recipients. These data indicate that defective DDR and diminished apoptotic priming provide a selective advantage to old HSCs that may contribute to mutation accrual and disease predisposition.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig 1: Old HSCs have increased survival upon DNA damage induction by a broad array of genotoxic agents.
Fig. 2: The function and viability of young and old HSCs are differentially affected by DNA damage induction.
Fig. 3: HSCs are the least apoptotically primed primitive progenitor population and exhibit differential dependence on apoptotic regulators during ageing.
Fig. 4: Apoptotic priming and survival upon DNA damage induction in HSC subsets.
Fig. 5: Apoptotic priming and survival upon DNA damage induction are equalized upon transplantation.
Fig. 6: ATM activity is reduced in old HSCs and leads to a diminished apoptotic priming response.

Similar content being viewed by others

References

  1. Rossi, D. J., Jamieson, C. H. & Weissman, I. L. Stems cells and the pathways to aging and cancer. Cell 132, 681–696 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Geiger, H., de Haan, G. & Florian, M. C. The ageing haematopoietic stem cell compartment. Nat. Rev. Immunol. 13, 376–389 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Beerman, I., Maloney, W. J., Weissmann, I. L. & Rossi, D. J. Stem cells and the aging hematopoietic system. Curr. Opin. Immunol. 22, 500–506 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Beerman, I., Seita, J., Inlay, M. A., Weissman, I. L. & Rossi, D. J. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle. Cell Stem Cell 15, 37–50 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Nijnik, A. et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Walter, D. et al. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature 520, 549–552 (2015).

    Article  PubMed  Google Scholar 

  8. Rube, C. E. et al. Accumulation of DNA damage in hematopoietic stem and progenitor cells during human aging. PLoS ONE 6, e17487 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Rossi, D. J. et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc. Natl Acad. Sci. USA 102, 9194–9199 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chambers, S. M. et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Beerman, I. et al. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12, 413–425 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Sun, D. et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Florian, M. C. et al. Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation. Cell Stem Cell 10, 520–530 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Beerman, I. et al. Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion. Proc. Natl Acad. Sci. USA 107, 5465–5470 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kim, M., Moon, H. B. & Spangrude, G. J. Major age-related changes of mouse hematopoietic stem/progenitor cells. Ann. NY Acad. Sci. 996, 195–208 (2003).

    Article  PubMed  Google Scholar 

  16. Morrison, S. J., Wandycz, A. M., Akashi, K., Globerson, A. & Weissman, I. L. The aging of hematopoietic stem cells. Nat. Med. 2, 1011–1016 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-associated characteristics of murine hematopoietic stem cells. J. Exp. Med. 192, 1273–1280 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Baguley, B. C. & Kerr, D. J. Anticancer drug development. Brit. J. Cancer 86, 1665–1666 (2002).

    Article  Google Scholar 

  19. Brunelle, J. K. & Letai, A. Control of mitochondrial apoptosis by the Bcl-2 family. J. Cell. Sci. 122, 437–441 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cory, S. & Adams, J. M. The Bcl2 family: regulators of the cellular life-or-death switch. Nat. Rev. Cancer 2, 647–656 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Llambi, F. et al. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol. Cell. 44, 517–531 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Letai, A. et al. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2, 183–192 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Ren, D. et al. BID, BIM, and PUMA are essential for activation of the BAX- and BAK-dependent cell death program. Science 330, 1390–1393 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hockenbery, D., Nunez, G., Milliman, C., Schreiber, R. D. & Korsmeyer, S. J. Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 348, 334–336 (1990).

    Article  CAS  PubMed  Google Scholar 

  25. Oda, E. et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053–1058 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Boyd, J. M. et al. Bik, a novel death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and interacts with viral and cellular survival-promoting proteins. Oncogene 11, 1921–1928 (1995).

    CAS  PubMed  Google Scholar 

  27. Kutuk, O. & Letai, A. Regulation of Bcl-2 family proteins by posttranslational modifications. Curr. Mol. Med. 8, 102–118 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Del Gaizo Moore, V. & Letai, A. BH3 profiling—measuring integrated function of the mitochondrial apoptotic pathway to predict cell fate decisions. Cancer Lett. 332, 202–205 (2013).

    Article  PubMed  Google Scholar 

  29. Ni Chonghaile, T. et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science 334, 1129–1133 (2011).

    Article  PubMed  Google Scholar 

  30. Ryan, J. A., Brunelle, J. K. & Letai, A. Heightened mitochondrial priming is the basis for apoptotic hypersensitivity of CD4+ CD8+ thymocytes. Proc. Natl Acad. Sci. USA 107, 12895–12900 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Vo, T. T. et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell 151, 344–355 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sarosiek, K. A. et al. Developmental regulation of mitochondrial apoptosis by c-Myc governs age- and tissue-specific sensitivity to cancer therapeutics. Cancer Cell 31, 142–156 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Sarosiek, K. A. et al. BID preferentially activates BAK while BIM preferentially activates BAX, affecting chemotherapy response. Mol. Cell 51, 751–765 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Opferman, J. T. et al. Obligate role of anti-apoptotic MCL-1 in the survival of hematopoietic stem cells. Science 307, 1101–1104 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Campbell, C. J. et al. The human stem cell hierarchy is defined by a functional dependence on Mcl-1 for self-renewal capacity. Blood 116, 1433–1442 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Villunger, A. et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins Puma and Noxa. Science 302, 1036–1038 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Dykstra, B., Olthof, S., Schreuder, J., Ritsema, M. & de Haan, G. Clonal analysis reveals multiple functional defects of aged murine hematopoietic stem cells. J. Exp. Med. 208, 2691–2703 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Muller-Sieburg, C. E., Cho, R. H., Karlsson, L., Huang, J. F. & Sieburg, H. B. Myeloid-biased hematopoietic stem cells have extensive self-renewal capacity but generate diminished lymphoid progeny with impaired IL-7 responsiveness. Blood 103, 4111–4118 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Benz, C. et al. Hematopoietic stem cell subtypes expand differentially during development and display distinct lymphopoietic programs. Cell Stem Cell 10, 273–283 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Challen, G. A., Boles, N. C., Chambers, S. M. & Goodell, M. A. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-β1. Cell Stem Cell 6, 265–278 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Morita, Y., Ema, H. & Nakauchi, H. Heterogeneity and hierarchy within the most primitive hematopoietic stem cell compartment. J. Exp. Med. 207, 1173–1182 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Beerman, I. & Rossi, D. J. Epigenetic control of stem cell potential during homeostasis, aging, and disease. Cell Stem Cell 16, 613–625 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Seita, J. et al. Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS ONE 7, e40321 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lukas, C. et al. Mdc1 couples DNA double-strand break recognition by Nbs1 with its H2AX-dependent chromatin retention. EMBO J. 23, 2674–2683 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bakkenist, C. J. & Kastan, M. B. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 421, 499–506 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. D’Amours, D. & Jackson, S. P. The Mre11 complex: at the crossroads of DNA repair and checkpoint signalling. Nat. Rev. Mol. Cell Biol. 3, 317–327 (2002).

    Article  PubMed  Google Scholar 

  47. Ito, K. et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431, 997–1002 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Maryanovich, M. et al. The ATM–BID pathway regulates quiescence and survival of haematopoietic stem cells. Nat. Cell Biol 14, 535–541 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Burma, S., Chen, B. P., Murphy, M., Kurimasa, A. & Chen, D. J. ATM phosphorylates histone H2AX in response to DNA double-strand breaks. J. Biol. Chem. 276, 42462–42467 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Flach, J. et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 512, 198–202 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schultz, L. B., Chehab, N. H., Malikzay, A. & Halazonetis, T. D. p53 binding protein 1 (53BP1) is an early participant in the cellular response to DNA double-strand breaks. J. Cell. Biol. 151, 1381–1390 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hickson, I. et al. Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res. 64, 9152–9159 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Bondar, T. & Medzhitov, R. p53-mediated hematopoietic stem and progenitor cell competition. Cell Stem Cell 6, 309–322 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Pang, W. W. et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc. Natl Acad. Sci. USA 108, 20012–20017 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jan, M. et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci. Transl. Med. 4, 149ra118 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Ley, T. J. et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 363, 2424–2433 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Busque, L. et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 44, 1179–1181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ryan, J. & Letai, A. BH3 profiling in whole cells by flourimeter or FACS. Methods 61, 156–164 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Shlevkov for help with statistics, U. Rajarajacholan for help with dot blots and all the members of the D.J.R. laboratory for help. The A.N. laboratory was supported by the Intramural Research Program of the NIH, the National Cancer Institute, the Center for Cancer Research and the Alex Lemonade Stand Foundation Award. L.H. was supported by the NIH fellowship F31CA186301. The A.L. laboratory was supported by the NIH grant P01 CA066996 and the Leukemia and Lymphoma Society Grant TRP6387-13. D.J.R. is supported by grants from the NIH (RO1HL107630, R00AG029760 and UO1DK072473-01) as well as grants from The Leona M. and Harry B. Helmsley Charitable Trust, The New York Stem Cell Foundation, The Harvard Stem Cell Institute and the American Federation for Aging Research.

Author information

Authors and Affiliations

Authors

Contributions

P.G.-M. designed, performed and analysed most of the experiments and wrote the manuscript. L.H. designed and performed the BH3 profiling experiments. M.N. provided technical help. M.K. performed the immunostainings. I.B. performed the comet assays, transplantation of the CD150 HSC subsets and provided critical guidance. R.S. and K.S. provided Bax–/– and Bak–/– mice. A.N. provided Atm–/– mice and ATM guidance. A.L. and D.J.R. designed the experiments and wrote the manuscript.

Corresponding authors

Correspondence to Anthony Letai or Derrick J. Rossi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Inclusion of CD48 did not alter purity of HSCs gated on LSKFlk2-CD34- in either young or old mice.

a) Quantification and b) representative flow plots of CD48 expression levels in young and old HSCs (LSKFlk2-CD34-/lo), MPP1s (LSKFlk2-CD34+) and MPP2s (LSKFlk2+CD34+). Source data are included in Supplementary Table 1.

Supplementary Figure 2 Clonal colony size is similarly affected in young and old HSCs upon DNA damage induction.

a) Representative FACS plots showing HSC gating strategy and post-sort purity from young and old mice. b) Representative cell cycle profile and quantification of G0/G1 and S/G2M in young and old HSCs and steady state and upon 18 hours of culture. Data pooled from 4 independent experiments. (yHSC n = 2, oHSC n = 4, yHSC 18h n = 2, oHSC 18h n = 5) c) Frequency of live young and old HSCs that have divided at the indicated days. Data pooled from 6 independent experiments. d–g) Quantification of colony size upon exposure to the indicated treatments at day 10 measured as diameter of the colony in d, e) young and old HSCs and f, g) young and old MPs. Gamma irradiation (IR), ethyl-nitrosourea (ENU), ethyl-methanesulfonate (EMS), hydroxyurea (HU) doxorubicin (Doxo). Cell numbers (n) between 7–490, exact cell number per condition indicated in Supplementary Table 1. For d–g) each dot represents individual cells, data pooled from 2 independent experiments. P<0.05 (*), P<0.005 (**), P<0.0005 (***). Two-tailed Student’s t-test. Centre bar represents mean and error bars represent SEM. Source data are included in Supplementary Table 1.

Supplementary Figure 3 Clonal colony survival and growth dynamics is differentially affected in young and old HSCs upon DNA damage induction.

Cell number and viability of single untreated or irradiated young and old HSCs during 8 days of clonal culturing. Each column depicts a day and each row indicates a cell. 288 single HSCs per condition were analysed.

Supplementary Figure 4 BH3 profiling in stem and progenitor cells.

a) Dots plots and histograms showing gating strategy and TMRE intensity for each of the indicated populations in young and old bone marrow in response to BIM 3 μM and 8 μM. CLP: common lymphoid progenitors, MP: myeloid progenitors, LSK: lineage-Sca1+c-kit+, GMP: granulocyte monocyte progenitor, CMP: common myeloid progenitor, MEP: megakaryocyte erythroid progenitor, MPP1: multi-potent progenitor 1, MPP2: multi-potent progenitor 2, HSC: haematopoietic stem cells. b, c) Apoptotic priming of Bax-/- and Bak-/- HSCs in response to b) BIM 80 μM and c) BID 80 μM. Each dot represents an individual mouse (n = 3-5). Data pooled from 4 independent experiments. d) Clonal survival of young and old Bax-/- and Bak-/- HSCs measured as a percentage of viable clones in response to IR. Each dot represents percent survival of 24 to 48 single HSCs (LSK CD34-/lo Flk2- and LSK CD34-/lo CD150+) purified from individual mice (n = 2–4 mice per group). Numbers above the graphs indicate total number of surviving clones (black) vs total number of clones analysed (grey). Data pooled from 4 independent experiments. e) Apoptotic priming of young and old MPs in response to BAD 80 μM, HRK 80 μM and NOXA 80 μM. Each dot represents individual mice. Data pooled from 2 independent experiments. f) Frequency of CD150 HSCs subsets in young and old bone marrow (n = 5). Data pooled from 6 independent experiments. g) Apoptotic priming of young and old HSC subsets in response to BIM (8 μM) and BID (3 μM). Same data as in Fig 3f. Numbers above the graphs indicate total number of surviving clones (black) vs total number of clones analysed (grey). *, P< 0.05, **, P< 0.005, ***, P< 0.0005 (Two-tailed Student’s t-test). Centre bar represents mean and error bars represent SEM. Source data are included in Supplementary Table 1.

Supplementary Figure 5 Apoptotic priming and survival upon DNA damage induction is equalized upon transplantation.

a-b) Non-competitive transplantation of 5x106 young and old bone marrow showing a) peripheral blood chimaerism of recipient mice over the time course of the experiment, and b) lineage contribution of donor cells in recipients shown at week 16. Data from 1 experiment, n = 9 individual young recipient mice. c) Apoptotic priming of MP in response to BIM (8 μM), BID (3 μM) and BAD (80 μM) in steady-state young and old bone marrow and in bone marrow transplant-derived young and old LSK and MP (21–23 weeks post-transplantation). Data pooled from 5 independent experiments, each dot represents an individual mouse (n = 3, 4 or 5 mice per group). d-e) Non-competitive transplantation of 5×106 young bone marrow showing d) peripheral blood chimaerism of recipient mice over the time course of the experiment and e) lineage contribution of donor cells in recipients shown at week 16. Data from 1 experiment, n = 5 individual 17- month old recipient mice. ***, P< 0.0005 (Two-tailed Student’s t-test). Centre bar represents mean and error bars represent SEM. Source data are included in Supplementary Table 1.

Supplementary Figure 6 ATM levels and activity are diminished in old HSCs.

a-b) Expression of a) DNA damage response and b) DNA damage sensor genes in young and old HSCs, (n = 3–5 mice per group, as indicated with dots). Data represents normalized signal intensity (data from Gene Expression Commons4,43, Gene Expression Omnibus (Series GSE55525). c) Olive tail moment of young and old freshly isolated HSCs or upon 1 hour in culture post irradiation with 2Gy. Data represent 1 experiment, n numbers represent cells and are indicated in the Figure. d) Dot blot showing γH2AX and actin levels in LSKs non-treated or irradiated (1 hour), with and without ATM inhibitor (ATMi, 10 μM). Experiment was performed once. e) Quantification of γH2AX levels in c-kit enriched bone marrow upon irradiation and 4h of culture with or ATMi. n = 3 mice. *, P< 0.05, **, P< 0.005, ***, P< 0.0005 (Two-tailed Student’s t-test). Centre bar represents mean and error bars represent SEM. Source data are included in Supplementary Table 1.

Supplementary Figure 7 Unprocessed dot blots for Supplementary Figure 6d.

Please note that in blot 1 the samples for NT 25 μM and 2Gy 25 μM are switched and do not correspond to the labels and NT 50 μM and 2Gy 50 μΜ samples were not treated with ATMi (KU55933). Experiment performed once.

Supplementary information

Supplementary Information

Supplementary Figures 1–7 and Supplementary Table legend.

Life Sciences Reporting Summary

Supplementary Table 1

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gutierrez-Martinez, P., Hogdal, L., Nagai, M. et al. Diminished apoptotic priming and ATM signalling confer a survival advantage onto aged haematopoietic stem cells in response to DNA damage. Nat Cell Biol 20, 413–421 (2018). https://doi.org/10.1038/s41556-018-0054-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0054-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing