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:

Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function

Abstract

DNA damage contributes to the aging of hematopoietic stem cells (HSCs), yet the underlying molecular mechanisms are not fully understood. In this study, we identified a heterogeneous functional role of microcephalin (MCPH1) in the nucleus and cytoplasm of mouse HSCs. In the nucleus, MCPH1 maintains genomic stability, whereas in the cytoplasm, it prevents necroptosis by binding with p-RIPK3. Aging triggers MCPH1 translocation from cytosol to nucleus, reducing its cytoplasmic retention and leading to the activation of necroptosis and deterioration of HSC function. Mechanistically, we found that KAT7-mediated lysine acetylation within the NLS motif of MCPH1 in response to DNA damage facilitates its nuclear translocation. Targeted mutation of these lysines inhibits MCPH1 translocation and, consequently, compromises necroptosis. The dysfunction of necroptosis signaling, in turn, improves the function of aged HSCs. In summary, our findings demonstrate that DNA damage-induced redistribution of MCPH1 promotes HSC aging and could have broader implications for aging and aging-related diseases.

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: Dysfunction of Mcph1 mildly disturbs hematopoietic homeostasis.
Fig. 2: Mcph1−/− HSCs exhibit accumulated DNA damage and severely compromised reconstitution capacity.
Fig. 3: MCPH1 antagonizes necroptosis by interacting with RIPK3.
Fig. 4: Dysfunction of RIPK3 cannot rescue DNA damage accumulation in HSCs upon Mcph1 deletion.
Fig. 5: MCPH1 translocates to the nucleus in response to DNA damage.
Fig. 6: KAT7 mediates MCPH1 nuclear translocation by direct acetylation of MCPH1 in response to DNA damage.
Fig. 7: HSCs are susceptible to necroptosis during aging.

Similar content being viewed by others

Data availability

RNA-seq data are deposited in the Gene Expression Omnibus under accession code GSE217101. Proteomics data from mass spectrometry are deposited in the ProteomeXchange Consortium through iProX65 under dataset identifier PXD048580. Further information and requests for resources and reagents should be directed to and will be fulfilled by J.W.

References

  1. de Haan, G. & Lazare, S. S. Aging of hematopoietic stem cells. Blood 131, 479–487 (2018).

    Article  PubMed  Google Scholar 

  2. 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  PubMed  Google Scholar 

  3. 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 

  4. 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 

  5. Mohrin, M. et al. Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell 7, 174–185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Schumacher, B., Pothof, J., Vijg, J. & Hoeijmakers, J. H. J. The central role of DNA damage in the ageing process. Nature 592, 695–703 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. McNeely, T., Leone, M., Yanai, H. & Beerman, I. DNA damage in aging, the stem cell perspective. Hum. Genet. 139, 309–331 (2020).

    Article  PubMed  Google Scholar 

  8. Tiwari, V. & Wilson, D. M. DNA damage and associated DNA repair defects in disease and premature aging. Am. J. Hum. Genet. 105, 237–257 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, J., Lu, X., Sakk, V., Klein, C. A. & Rudolph, K. L. Senescence and apoptosis block hematopoietic activation of quiescent hematopoietic stem cells with short telomeres. Blood 124, 3237–3240 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lin, S.-Y. & Elledge, S. J. Multiple tumor suppressor pathways negatively regulate telomerase. Cell 113, 881–889 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Jackson, A. P. et al. Identification of microcephalin, a protein implicated in determining the size of the human brain. Am. J. Hum. Genet. 71, 136–142 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gruber, R. et al. MCPH1 regulates the neuroprogenitor division mode by coupling the centrosomal cycle with mitotic entry through the Chk1–Cdc25 pathway. Nat. Cell Biol. 13, 1325–1334 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Liang, Y. et al. BRIT1/MCPH1 is essential for mitotic and meiotic recombination DNA repair and maintaining genomic stability in mice. PLoS Genet. 6, e1000826 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Lin, S. Y., Rai, R., Li, K., Xu, Z. X. & Elledge, S. J. BRIT1/MCPH1 is a DNA damage responsive protein that regulates the Brca1-Chk1 pathway, implicating checkpoint dysfunction in microcephaly. Proc. Natl Acad. Sci. USA 102, 15105–15109 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Peng, G. et al. BRIT1/MCPH1 links chromatin remodelling to DNA damage response. Nat. Cell Biol. 11, 865–872 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Alderton, G. K. et al. Regulation of mitotic entry by microcephalin and its overlap with ATR signalling. Nat. Cell Biol. 8, 725–733 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Tibelius, A. et al. Microcephalin and pericentrin regulate mitotic entry via centrosome-associated Chk1. J. Cell Biol. 185, 1149–1157 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Brown, J. A. L., Bourke, E., Liptrot, C., Dockery, P. & Morrison, C. G. MCPH1/BRIT1 limits ionizing radiation-induced centrosome amplification. Oncogene 29, 5537–5544 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Journiac, N. et al. Cell metabolic alterations due to Mcph1 mutation in microcephaly. Cell Rep. 31, 107506 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Wood, J. L., Liang, Y., Li, K. & Chen, J. Microcephalin/MCPH1 associates with the condensin II complex to function in homologous recombination repair. J. Biol. Chem. 283, 29586–29592 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wu, X. et al. Microcephalin regulates BRCA2 and Rad51-associated DNA double-strand break repair. Cancer Res. 69, 5531–5536 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rai, R. et al. BRIT1 regulates early DNA damage response, chromosomal integrity, and cancer. Cancer Cell 10, 145–157 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu, X. et al. The N-terminal BRCT domain determines MCPH1 function in brain development and fertility. Cell Death Dis. 12, 143 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jeffers, L. J., Coull, B. J., Stack, S. J. & Morrison, C. G. Distinct BRCT domains in Mcph1/Brit1 mediate ionizing radiation-induced focus formation and centrosomal localization. Oncogene 27, 139–144 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Xu, X., Lee, J. & Stern, D. F. Microcephalin is a DNA damage response protein involved in regulation of CHK1 and BRCA1. J. Biol. Chem. 279, 34091–34094 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Ling, J. P. et al. ASCOT identifies key regulators of neuronal subtype-specific splicing. Nat. Commun. 11, 137 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liu, X., Zhou, Z. W. & Wang, Z. Q. The DNA damage response molecule MCPH1 in brain development and beyond. Acta Biochim. Biophys. Sin. 48, 678–685 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Wang, J. et al. A differentiation checkpoint limits hematopoietic stem cell self-renewal in response to DNA damage. Cell 148, 1001–1014 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Koulnis, M. et al. Identification and analysis of mouse erythroid progenitors using the CD71/TER119 flow-cytometric assay. J. Vis. Exp. https://doi.org/10.3791/2809 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Liu, M., Li, H., Yang, R., Ji, D. & Xia, X. GSK872 and necrostatin-1 protect retinal ganglion cells against necroptosis through inhibition of RIP1/RIP3/MLKL pathway in glutamate-induced retinal excitotoxic model of glaucoma. J. Neuroinflammation 19, 262 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. He, S. et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 137, 1100–1111 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Yu, X., Chini, C. C. S., He, M., Mer, G. & Chen, J. The BRCT domain is a phospho-protein binding domain. Science 302, 639–642 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Chen, W. et al. Diverse sequence determinants control human and mouse receptor interacting protein 3 (RIP3) and mixed lineage kinase domain-like (MLKL) interaction in necroptotic signaling. J. Biol. Chem. 288, 16247–16261 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xie, Y. et al. Gut epithelial TSC1/mTOR controls RIPK3-dependent necroptosis in intestinal inflammation and cancer. J. Clin. Invest. 130, 2111–2128 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Lange, A. et al. Classical nuclear localization signals: definition, function, and interaction with importin α. J. Biol. Chem. 282, 5101–5105 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Horton, P. et al. WoLF PSORT: protein localization predictor. Nucleic Acids Res. 35, W585–W587 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Cao, X. et al. Acetylation promotes TyrRS nuclear translocation to prevent oxidative damage. Proc. Natl Acad. Sci. USA 114, 687–692 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Narita, T., Weinert, B. T. & Choudhary, C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 20, 156–174 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. MacPherson, L. et al. HBO1 is required for the maintenance of leukaemia stem cells. Nature 577, 266–270 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Newton, K., Sun, X. Q. & Dixit, V. M. Kinase RIP3 is dispensable for normal NF-KBs, signaling by the B-cell and T-cell receptors, tumor necrosis factor receptor 1, and toll-like receptors 2 and 4. Mol. Cell. Biol. 24, 1464–1469 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li, D. et al. RIPK1-RIPK3-MLKL-dependent necrosis promotes the aging of mouse male reproductive system. Elife 6, e27692 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Leist, M., Single, B., Castoldi, A. F., Kühnle, S. & Nicotera, P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J. Exp. Med. 185, 1481–1486 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ferrari, D., Stepczynska, A., Los, M., Wesselborg, S. & Schulze-Osthoff, K. Differential regulation and ATP requirement for caspase-8 and caspase-3 activation during CD95- and anticancer drug–induced apoptosis. J. Exp. Med. 188, 979–984 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Elmore, S. Apoptosis: a review of programmed cell death. Toxicol. Pathol. 35, 495–516 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Eguchi, Y., Shimizu, S. & Tsujimoto, Y. Intracellular ATP levels determine cell death fate by apoptosis or necrosis. Cancer Res. 57, 1835–1840 (1997).

    CAS  PubMed  Google Scholar 

  47. Nombela-Arrieta, C. et al. Quantitative imaging of haematopoietic stem and progenitor cell localization and hypoxic status in the bone marrow microenvironment. Nat. Cell Biol. 15, 533–543 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Takubo, K. et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Nakamura-Ishizu, A., Ito, K. & Suda, T. Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Benayoun, B. A., Pollina, E. A. & Brunet, A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat. Rev. Mol. Cell Biol. 16, 593–610 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schwer, B. et al. Calorie restriction alters mitochondrial protein acetylation. Aging Cell 8, 604–606 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Newman, J. C. et al. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell metabolism 26, 547–557 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Roberts, M. N. et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell metabolism 26, 539–546 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Klein, H.-U. et al. Epigenome-wide study uncovers large-scale changes in histone acetylation driven by tau pathology in aging and Alzheimer’s human brains. Nat. Neurosci. 22, 37–46 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Krishnan, V. et al. Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24-deficient mice. Proc. Natl Acad. Sci. USA 108, 12325–12330 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Peleg, S. et al. Altered histone acetylation is associated with age-dependent memory impairment in mice. Science 328, 753–756 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Peleg, S. et al. Lifespan extension by targeting a link between metabolism and histone acetylation in Drosophila. Embo Rep. 17, 455–469 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wang, W. et al. A genome-wide CRISPR-based screen identifies KAT7 as a driver of cellular senescence. Sci. Transl. Med. 13, eabd2655 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Yang, Y. et al. The histone lysine acetyltransferase HBO1 (KAT7) regulates hematopoietic stem cell quiescence and self-renewal. Blood 139, 845–858 (2022).

    Article  CAS  PubMed  Google Scholar 

  60. Imai, S. -I. & Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 24, 464–471 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hall, J. A., Dominy, J. E., Lee, Y. & Puigserver, P. The sirtuin family’s role in aging and age-associated pathologies. J. Clin. Invest. 123, 973–979 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. He, H., Wang, Y. & Wang, J. ALKBH3 is dispensable in maintaining hematopoietic stem cells but forced ALKBH3 rectified the differentiation skewing of aged hematopoietic stem cells. Blood Sci. 2, 137–143 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. He, H. et al. Aging-induced IL27Ra signaling impairs hematopoietic stem cells. Blood 136, 183–198 (2020).

    Article  PubMed  Google Scholar 

  65. Chen, T. et al. iProX in 2021: connecting proteomics data sharing with big data. Nucleic Acids Res. 50, D1522–D1527 (2021).

    Article  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank X. Wang (National Institute of Biological Sciences) for providing Ripk3−/− mice. We thank G. -H. Liu (Institute of Zoology, Chinese Academy of Sciences) for providing Kat7 cDNA. We thank S. -J. Chen (Shanghai Jiao Tong University School of Medicine) for providing the 32D cell line. This work was supported by grants Z200022, 82250002, 92249305 and 2018YFA0800200 to J.W. from the National Key R&D Program of China or the Beijing Municipal Science & Technology Commission and the National Natural Science Foundation of China.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, J.W.; Methodology, H.H., Y.W., B.T., Q.D. and C.W.; Investigation, H.H., Y.W., B.T., Q.D., C.W. and W.S.; Formal Analysis, J.W. and H.H.; Resources, J.W.; Writing, J.W.; Funding acquisition, J.W.; Supervision, J.W.

Corresponding author

Correspondence to Jianwei Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Zhaoqi Wang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Dysfunction of Mcph1 mildly disturbs hematopoietic homeostasis at young age.

(a) This histogram shows the expression of MCPH1 in various immune cells. Data were download from ASCOT. (b) The schematic diagram showing the targeting strategy to generate Mcph1flox mice. (c) The schematic diagram showing the breeding strategy to generate Mcph1−/− (Vav1-Cre; Mcph1flox/flox) mice. (d-j) 2-5 months old Mcph1−/− or WT mice were analyzed for hematopoietic progenitor cells and HSCs. (d-e) The histogram depicts the frequency (d) and the absolute numbers (e) of HSCs (CD34- CD150+ KSL). n = 7 mice per group. (f-g) The histogram shows the frequency (f) and the absolute numbers (g) of CMPs, GMPs, and MEPs. n = 7 mice per group. (h-i) The histogram shows the frequency (h) and the absolute numbers (i) of CLPs. n = 4 mice per group. (j) The scatter plots show the bone marrow (BM) cellularity of Mcph1−/− and WT mice. n = 7 mice per group. (k-n) 3-4 months old Mcph1−/− or WT mice were analyzed for thymocyte. (k) Representative flow cytometry of thymocytes for CD4/CD8 expression. (l) The scatter plots show the total cell number of thymus. (m-n) The scatter plots depict the percentage (m) and absolute numbers (n) of DN (CD4CD8), DP (CD4+CD8+), CD4+, and CD8+ T cells in thymus. n = 6-7 mice per group (WT: n = 7; Mcph1−/−: n = 6). (o-r) 3-4 months old Mcph1−/− or WT mice were analyzed for splenocytes. (o) Representative flow cytometry. (p) The scatter plots show the total cell number of the spleen. (q-r) The scatter plots depict the percentage (q) and absolute numbers (r) of myeloid, B, CD4+ T and CD8+ T cells in spleen. n = 6-7 mice per group (WT: n = 7; Mcph1−/−: n = 6). Two-tailed unpaired Student’s t-test was used for statistical analysis in l-n and q-r. Data are shown as mean ± SD in d-j, l-n and p-r.

Source data

Extended Data Fig. 2 Mcph1 dysfunction severely impairs the reconstitution capacity of HSCs.

(a-f) Freshly isolated 20 HSCs from 4-month old Mcph1−/− mice or age-matched WT mice were transplanted into lethally irradiated recipients together with 3 × 105 competitor cells. Chimera in peripheral blood was checked every month until the 3rd month. (a) This histogram displays the percentage of donor cell reconstitution in overall (CD45.2+), B (B220+), T (CD3+) and myeloid (Mac-1+) cell every month after transplantation. (b and d) The scatter plots show the percentage of donor cell reconstitution in bone marrow (b) and HSC (d). (c) This histogram displays the lineage distribution in donor-derived bone marrow at the 3rd month. (e) This histogram displays the percentage of donor cell reconstitution in erythroid progenitors (Ter119+) every month after transplantation. (f) the scatter plots show the percentage of donor cell reconstitution in erythroid progenitors of bone marrow. n = 6-7 mice per group in a and e (n =6 for WT, n =7 for Mcph1−/−). n = 6 mice per group in b, c, d, and f. Data were shown as mean ± SD. Two-tailed unpaired Student’s t-test was used for statistical analysis.

Source data

Extended Data Fig. 3 MCPH1 antagonizes necroptosis through interaction with RIPK3.

(a-b) 4 months old Mcph1−/− or WT mice were analyzed for cell death of fresh HSCs without ex vivo culture by Annexin-V and DAPI staining. (a) Representative flow cytometry. (b) Percentage of early stage of cell death (DAPI Annexin-V+) in Mcph1−/− or WT HSCs. n = 4-5 mice per group (WT: n = 5; Mcph1−/−: n = 4). (c) The images show morphological alteration of WT and Mcph1−/− HSCs (3 months). The experiment was repeated 3 times. (d) Representative western blot shows the interaction between MCPH1 and RIPK3 in response to necroptotic stimuli. L929 cells stably expressing Flag-tagged MCPH1 were treated with T+Z for 1 hour. Cell lysates were immunoprecipitated with anti-Flag antibody, and western blot was performed with indicated antibodies. The experiment was repeated 3 times. (e) Interaction between MCPH1 and p-RIPK3 in HEK293T cells co-transfected with SFB-tagged MCPH1 and Myc-tagged RIPK3, demonstrated by co-immunoprecipitation using S beads and subsequent western blot analysis with specific antibodies. The experiment was repeated 3 times. (f) SFB-RIPK3 were transfected into HEK293T cells together with GFP-MLKL and Myc-MCPH1-WT or Myc-MCPH1-Δ624–882 constructs as indicated. Cells were subjected to immunoprecipitated with S beads and western blot analysis was performed with indicated antibodies. Representative western blot shows that MCPH1-WT but not MCPH1-Δ624-882 inhibits interaction between p-RIPK3 and MLKL. The experiment was repeated 3 times. (g) Co-immunoprecipitation assay in HEK293T cells transiently transfected with SFB- MCPH1 and either Myc-tagged wild-type or T231A-S232A RIPK3, revealing a reduced interaction between MCPH1 and the RIPK3T231A-S232A mutant, as indicated by western blot analysis. The experiment was repeated 3 times. (h) The mRNA expression of Ripk3 in aged (23 months) Mcph1−/− and WT HSCs. n = 3 independent experiments. (i) Representative western blot showing the expression of p-S6, S6, p-4EBP1, 4EBP1, and MCPH1 in aged (21–26 months) Mcph1−/− and WT HSCs. The experiment was repeated twice. Two-tailed unpaired Student’s t-test was used for statistical analysis in b. Data are shown as mean ± SD in b and h.

Source data

Extended Data Fig. 4 Dysfunction of Ripk3 partially rescues the deleterious impact of Mcph1 deletion on HSCs during short-term ex vivo culture.

(a-c) Freshly isolated KSL cells either from Ripk3−/− mice (2 months old) or age-matched WT mice were infected by lentivirus carrying Mcph1 shRNA or scramble control (shRNA expressing lentiviruses coexpressed GFP), and 72 hours later, FACS-purified 100 GFP+ CD48 Sca1+ cells were cultured in SFEM medium. 7 days later, the proliferation capacity was evaluated by examining the colony size and cell number. (a) Experimental design. (b) Representative images of ex vivo-cultured HSCs carrying the indicated shRNA on day 7. Scale bar, 50 μm. (c) The scatter plots depict the cell numbers of ex vivo-cultured HSCs carrying the indicated shRNA on day 7. Statistical significance was measured by two-way ANOVA followed by Sidak′s test for post hoc comparison. n = 18–20 independent experiments (n = 20 for WT: shCon, n = 19 for WT: shMcph1, n = 18 for Ripk3−/−: shCon, n = 20 for Ripk3−/−: shMcph1). Data were shown as mean±SD.

Source data

Extended Data Fig. 5 Nuclear translocation of MCPH1 in response to DNA damage.

(a-b) Cells were treated with 100 μM etoposide and 1 hour later, cell lysates were separated into cytoplasmic and nuclear fractions. The subcellular distribution of MCPH1 were analyzed by western blot. GAPDH and H3 were used to detect the purity of cytoplasmic and nuclear fractions respectively. (a) EL4 cells. (b) 32D cells. The experiment was repeated 3 times. (c) KSL cells were treated with 100 μM etoposide and 1 hour later, cell lysates were separated into cytoplasmic and nuclear fractions. The subcellular distribution of RIPK3 were analyzed by western blot. The experiment was repeated 3 times.

Source data

Extended Data Fig. 6 KAT7-dependent acetylation of MCPH1 facilitating nuclear translocation in response to DNA damage.

(a) HEK293T cells were either mock treated or treated with 100 μM etoposide with or without 1 μM KAT7 inhibitor for 1 hours. cell lysates were separated into cytoplasmic and nuclear fractions. The subcellular distribution of MCPH1 were analyzed by western blot. The experiment was repeated 3 times. (b) HEK293T cells were transiently transfected with SFB-tagged wild-type MCPH1 or indicated K/R mutants together with Myc-tagged KAT7. Cell lysates were immunoprecipitated with S beads, and western blot analysis was performed with indicated antibodies. Representative western blot shows the acetylation level of wild-type or various K/R mutants of MCPH1. The experiment was repeated twice. (c) HEK293T cells were transiently transfected with SFB-tagged wild-type MCPH1 or MCPH1K348/350R, MCPH1K360/362R, MCPH1K364/365R, MCPH1K360/362/364/365R together with Myc-tagged KAT7. Cell lysates were immunoprecipitated with S beads, and western blot analysis was performed with indicated antibodies. Representative western blot shows the acetylation level of wild-type or indicated mutants of MCPH1. The experiment was repeated 3 times. (d) HEK293T cells were transiently transfected with SFB-tagged wild-type MCPH1 or K348R-K350R-K360R-K362R mutant together with Myc-tagged KAT7. 24 hours later, cell lysates were immunoprecipitated with S beads, and western blot analysis was performed with indicated antibodies. Representative western blot shows the interaction between MCPH1K348/350/360/362R and MCPH1 with KAT7. The experiment was repeated 3 times. (e) HEK293T cells were transiently transfected with SFB-tagged wild-type MCPH1 or K348R-K350R-K360R-K362R mutant together with Myc-tagged RIPK3. 24 hours later, cell lysates were immunoprecipitated with S beads, and western blot analysis was performed with indicated antibodies. Representative western blot shows the interaction between MCPH1K348/350/360/362R and MCPH1 with RIPK3. The experiment was repeated twice. (f) MCPH1- and MCPH1K348R-K350R-K360R-K362R-expressed L929 cells were treated with etoposide + zVAD (100 μM etoposide, 20 μM zVAD) for 20 hours. Representative western blot shows the expression of p-MLKL, MLKL, p-RIPK3, RIPK3, and MCPH1 in WT or K348R-K350R-K360R-K362R mutant MCPH1 overexpressed cells upon etoposide + zVAD treatment. The experiment was repeated 3-4 times.

Source data

Extended Data Fig. 7 Enhanced susceptibility of HSCs to necroptosis during aging.

(a) The images show morphological alteration of in Mlkl−/− and WT HSCs at young (3 months) and old (29 months) age. (b) The scatter plots depict the percentage of necroptotic cell death in Mlkl−/− and WT HSCs at young (3 months) and old (29 months) age. Data were shown as mean ± SD. Statistical significance was measured by two-way ANOVA followed by Tukey’s test for post hoc comparison. n = 3 independent experiments. (c-f) KSL cells were freshly isolated from young (5 months) or old (18 months) mice were infected by lentivirus carrying the cDNA of WT MCPH1 or MCPH1- K348R-K350R-K360R-K362R mutant (MCPH14KR), and 72 hours later, 2 × 104 GFP+ cells were sorted and injected into lethally irradiated recipients together with 2 × 105 competitor cells. Peripheral blood of recipient mice was evaluated every month until the 4th month. This histogram displays the percentage of GFP+ cell in overall (CD45.2+), B (B220+), T (CD3+) and myeloid (Mac-1+) cell every month after transplantation. n = 6-7 mice per group (n = 6 for Young: Vector, Young: MCPH1-WT, Old: Vector, and Old: MCPH1-WT; n = 7 for Young: MCPH1-4KR and Old: MCPH1-4KR), data were shown as mean ± SD. Two-tailed unpaired Student’s t-test was used for statistical analysis.

Source data

Extended Data Fig. 8 Representative flow cytometry gating strategy used in this study.

(a) Representative flow cytometry showing the gating strategy used to isolate and analyze the hematopoietic stem and progenitor cell populations in mouse bone marrow presented on Figs. 1a,b,2a–h,3a–h,l,o,4a, b, j, k,5a, b,d–f,6n–o, F,7a–d, g–i, j, l and Extended Data Figs. 1d–h, 2a, d, 3a–c, h, i, 4a, b, 5c, 7a–f. (b) Strategy used to evaluate the the percentage of B, myeloid, CD4+ T, CD8+ T cells presented on Fig. 1d, e. (c-d) Strategy used to evaluate the the percentage (c) and lineage distribution (d) of test donor-derived cells (myeloid, B and T cells) presented on Fig. 2f–g, 4c–i, 7g, j, k, l, m, and Extended Data Figs. 2a–c, 7c–f. (e) Strategy used to evaluate the the percentage of test donor-derived erythroid progenitors (Ter119+) presented on Extended Data Fig. 2e, f.

Supplementary information

Supplementary Information

Working model

Reporting Summary

Supplementary Tables 1–3

Supplementary Table 1. GSEA gene list; Supplementary Table 2. Oligonucleotides; Supplementary Table 3. Materials;

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, H., Wang, Y., Tang, B. et al. Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function. Nat Aging 4, 510–526 (2024). https://doi.org/10.1038/s43587-024-00609-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-024-00609-z

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