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:

Transmembrane nuclease NUMEN/ENDOD1 regulates DNA repair pathway choice at the nuclear periphery

An Author Correction to this article was published on 01 August 2023

This article has been updated

Abstract

Proper repair of DNA damage lesions is essential to maintaining genome integrity and preventing the development of human diseases, including cancer. Increasing evidence suggests the importance of the nuclear envelope in the spatial regulation of DNA repair, although the mechanisms of such regulatory processes remain poorly defined. Through a genome-wide synthetic viability screen for PARP-inhibitor resistance using an inducible CRISPR–Cas9 platform and BRCA1-deficient breast cancer cells, we identified a transmembrane nuclease (renamed NUMEN) that could facilitate compartmentalized and non-homologous end joining-dependent repair of double-stranded DNA breaks at the nuclear periphery. Collectively, our data demonstrate that NUMEN generates short 5′ overhangs through its endonuclease and 3′→5′ exonuclease activities, promotes the repair of DNA lesions—including heterochromatic lamina-associated domain breaks as well as deprotected telomeres—and functions as a downstream effector of DNA-dependent protein kinase catalytic subunit. These findings underline the role of NUMEN as a key player in DNA repair pathway choice and genome-stability maintenance, and have implications for ongoing research into the development and treatment of genome instability disorders.

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: A genome-wide inducible CRISPR screen identifies NUMEN as a regulator of PARPi resistance in BRCA1-deficient tumour cells.
Fig. 2: NUMEN exhibits endonuclease as well as 3′→5′ exonuclease activities and generates DNA termini preferred by NHEJ repair.
Fig. 3: NUMEN antagonizes the formation of 3′ overhang in cells and regulates NHEJ/HR repair pathway choice.
Fig. 4: NUMEN is an important regulator of NHEJ and functions downstream of DNA-PKcs.
Fig. 5: NUMEN localizes to the nuclear membrane and spatially controls DNA damage response.
Fig. 6: NUMEN could help resolve perinuclear heterochromatic DSBs.
Fig. 7: NUMEN contributes to the maintenance of genomic stability.

Similar content being viewed by others

Data availability

Mass spectrometry data have been deposited in ProteomeXchange with the primary accession code PXD041562. Sequencing data from the CRISPR screen have been deposited in the NCBI Sequence Read Archive under the accession number PRJNA933943. The human breast cancer and pan-cancer data were derived from the TCGA Research Network (http://cancergenome.nih.gov/). The dataset derived from this resource that supports the findings of this study is available at https://github.com/Chenlt5/paper. The next-generation sequencing and mass spectrometric data have been deposited to the Figshare database and are available at https://doi.org/10.6084/m9.figshare.21592878. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

Custom codes are available at https://github.com/Chenlt5/paper.

Change history

References

  1. Khanna, K. K. & Jackson, S. P. DNA double-strand breaks: signaling, repair and the cancer connection. Nat. Genet. 27, 247–254 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Krenning, L., van den Berg, J. & Medema, R. H. Life or death after a break: what determines the choice?. Mol. Cell 76, 346–358 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Wright, W. D., Shah, S. S. & Heyer, W. Homologous recombination and the repair of DNA double-strand breaks. J. Biol. Chem. 293, 10524–10535 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sung, P. & Klein, H. Mechanism of homologous recombination: mediators and helicases take on regulatory functions. Nat. Rev. Mol. Cell Biol. 7, 739–750 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Stankiewicz, P. & Lupski, J. R. Genome architecture, rearrangements and genomic disorders. Trends Genet. 18, 74–82 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Lupski, J. R. Genomic disorders: structural features of the genome can lead to DNA rearrangements and human disease traits. Trends Genet. 14, 417–422 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Lieber, M. R., Ma, Y., Pannicke, U. & Schwarz, K. Mechanism and regulation of human non-homologous DNA end-joining. Nat. Rev. Mol. Cell Biol 4, 712–720 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Ghosh, D. & Raghavan, S. C. Nonhomologous end joining: new accessory factors fine tune the machinery. Trends Genet. 37, 582–599 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Weterings, E. & Chen, D. J. The endless tale of non-homologous end-joining. Cell Res. 18, 114–124 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Chang, H. H. Y. et al. Different DNA end configurations dictate which NHEJ components are most important for joining efficiency. J. Biol. Chem. 291, 24377–24389 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Scully, R., Panday, A., Elango, R. & Willis, N. A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 20, 698–714 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liang, Z., Sunder, S., Nallasivam, S. & Wilson, T. E. Overhang polarity of chromosomal double-strand breaks impacts kinetics and fidelity of yeast non-homologous end joining. Nucleic Acids Res. 44, 2769–2781 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ceccaldi, R., Rondinelli, B. & D Andrea, A. D. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 26, 52–64 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Lemaître, C. et al. Nuclear position dictates DNA repair pathway choice. Gene Dev. 28, 2450–2463 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Oza, P., Jaspersen, S. L., Miele, A., Dekker, J. & Peterson, C. L. Mechanisms that regulate localization of a DNA double-strand break to the nuclear periphery. Gene Dev. 23, 912–927 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tsouroula, K. et al. Temporal and spatial uncoupling of DNA double strand break repair pathways within mammalian heterochromatin. Mol. Cell 63, 293–305 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Goodarzi, A. A., Noon, A. T. & Jeggo, P. A. The impact of heterochromatin on DSB repair. Biochem. Soc. Trans. 37, 569–576 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Fontana, G. A. et al. Rif1 S-acylation mediates DNA double-strand break repair at the inner nuclear membrane. Nat. Commun. 10, 2535 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Nagai, S. et al. Functional targeting of DNA damage to a nuclear pore-associated SUMO-dependent ubiquitin ligase. Science 322, 597–602 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Goodarzi, A. A. et al. ATM signaling facilitates repair of DNA double-strand breaks associated with heterochromatin. Mol. Cell 31, 167–177 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Li, L., Guan, Y., Chen, X., Yang, J. & Cheng, Y. DNA repair pathways in cancer therapy and resistance. Front. Pharm. 11, 629266 (2020).

    Article  CAS  Google Scholar 

  22. Trenner, A. & Sartori, A. A. Harnessing DNA double-strand break repair for cancer treatment. Front. Oncol. 9, 1388 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Pilié, P. G., Tang, C., Mills, G. B. & Yap, T. A. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat. Rev. Clin. Oncol. 16, 81–104 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. D’Andrea, A. D. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair 71, 172–176 (2018).

    Article  PubMed  Google Scholar 

  25. Rose, M., Burgess, J. T., O’Byrne, K., Richard, D. J. & Bolderson, E. PARP Inhibitors: clinical relevance, mechanisms of action and tumor resistance. Front Cell Dev. Biol. 8, 564601 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Noordermeer, S. M. & van Attikum, H. PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells. Trends Cell Biol. 29, 820–834 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Li, H. et al. PARP inhibitor resistance: the underlying mechanisms and clinical implications. Mol. Cancer 19, 107 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kim, H. et al. Systematic analysis of human telomeric dysfunction using inducible telosome/shelterin CRISPR/Cas9 knockout cells. Cell Discov. 3, 17034 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bunting, S. F. et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 141, 243–254 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Xu, G. et al. REV7 counteracts DNA double-strand break resection and affects PARP inhibition. Nature 521, 541–544 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Mateo, J. et al. A decade of clinical development of PARP inhibitors in perspective. Ann. Oncol. 30, 1437–1447 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gupta, R. et al. DNA repair network analysis reveals shieldin as a key regulator of NHEJ and PARP inhibitor sensitivity. Cell 173, 972–988 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chang, H. H. Y., Pannunzio, N. R., Adachi, N. & Lieber, M. R. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat. Rev. Mol. Cell Biol. 18, 495–506 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Pannunzio, N. R., Watanabe, G. & Lieber, M. R. Nonhomologous DNA end-joining for repair of DNA double-strand breaks. J. Biol. Chem. 293, 10512–10523 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Lin, J. L. J., Wu, C., Yang, W. & Yuan, H. S. Crystal structure of endonuclease G in complex with DNA reveals how it nonspecifically degrades DNA as a homodimer. Nucleic Acids Res. 44, 10480–10490 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Cymerman, I. A., Chung, I., Beckmann, B. M., Bujnicki, J. M. & Meiss, G. EXOG, a novel paralog of endonuclease G in higher eukaryotes. Nucleic Acids Res. 36, 1369–1379 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhao, F. et al. ASTE1 promotes shieldin-complex-mediated DNA repair by attenuating end resection. Nat. Cell Biol. 23, 894–904 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Ashley, A. K. et al. DNA-PK phosphorylation of RPA32 Ser4/Ser8 regulates replication stress checkpoint activation, fork restart, homologous recombination and mitotic catastrophe. DNA Repair 21, 131–139 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, Y., Caron, P., Legube, G. & Paull, T. T. Quantitation of DNA double-strand break resection intermediates in human cells. Nucleic Acids Res. 42, e19 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Bennardo, N., Gunn, A., Cheng, A., Hasty, P. & Stark, J. M. Limiting the persistence of a chromosome break diminishes its mutagenic potential. PLoS Genet. 5, e1000683 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Riballo, E. et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to γ-H2AX foci. Mol. Cell 16, 715–724 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Noon, A. T. et al. 53BP1-dependent robust localized KAP-1 phosphorylation is essential for heterochromatic DNA double-strand break repair. Nat. Cell Biol. 12, 177–184 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Chapman, J. R. et al. RIF1 is essential for 53BP1-dependent nonhomologous end joining and suppression of DNA double-strand break resection. Mol. Cell 49, 858–871 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Celli, G. B. & de Lange, T. DNA processing is not required for ATM-mediated telomere damage response after TRF2 deletion. Nat. Cell Biol. 7, 712–718 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Branon, T. C. et al. Efficient proximity labeling in living cells and organisms with TurboID. Nat. Biotechnol. 36, 880–887 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kind, J. et al. Genome-wide maps of nuclear lamina interactions in single human cells. Cell 163, 134–147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Janin, A., Bauer, D., Ratti, F., Millat, G. & Méjat, A. Nuclear envelopathies: a complex LINC between nuclear envelope and pathology. Orphanet J. Rare Dis. 12, 147 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Falck, J., Coates, J. & Jackson, S. P. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature 434, 605–611 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Lemaitre, C. & Soutoglou, E. DSB (im)mobility and DNA repair compartmentalization in mammalian cells. J. Mol. Biol. 427, 652–658 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Horigome, C. et al. SWR1 and INO80 chromatin remodelers contribute to DNA double-strand break perinuclear anchorage site choice. Mol. Cell 55, 626–639 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Misteli, T. & Soutoglou, E. The emerging role of nuclear architecture in DNA repair and genome maintenance. Nat. Rev. Mol. Cell Biol. 10, 243–254 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Fortuny, A. & Polo, S. E. The response to DNA damage in heterochromatin domains. Chromosoma 127, 291–300 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Povirk, L. F. DNA damage and mutagenesis by radiomimetic DNA-cleaving agents: bleomycin, neocarzinostatin and other enediynes. Mutat. Res. 355, 71–89 (1996).

    Article  PubMed  Google Scholar 

  57. Povirk, L. F., Goldar, A. & Greenwood, M. Processing of damaged DNA ends for double-strand break repair in mammalian cells. ISRN Mol. Biol. 2012, 345805 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Meister, P., Towbin, B. D., Pike, B. L., Ponti, A. & Gasser, S. M. The spatial dynamics of tissue-specific promoters during C. elegans development. Gene Dev. 24, 766–782 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yang, K. S., Kohler, R. H., Landon, M., Giedt, R. & Weissleder, R. Single cell resolution in vivo imaging of DNA damage following PARP inhibition. Sci. Rep. 5, 10129 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kind, J. et al. Single-cell dynamics of genome-nuclear lamina interactions. Cell 153, 178–192 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Altemose, N. et al. μDamID: a microfluidic approach for joint imaging and sequencing of protein-dna interactions in single cells. Cell Syst. 11, 354–366 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. van Steensel, B. & Belmont, A. S. Lamina-associated domains: links with chromosome architecture, heterochromatin, and gene repression. Cell 169, 780–791 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Kemp, M. G. The histone deacetylase inhibitor trichostatin A alters the pattern of DNA replication origin activity in human cells. Nucleic Acids Res. 33, 325–336 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Meuleman, W. et al. Constitutive nuclear lamina-genome interactions are highly conserved and associated with A/T-rich sequence. Genome Res 23, 270–280 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Knijnenburg, T. A. et al. Genomic and molecular landscape of DNA damage repair deficiency across The Cancer Genome Atlas. Cell Rep. 23, 239–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Marquard, A. M. et al. Pan-cancer analysis of genomic scar signatures associated with homologous recombination deficiency suggests novel indications for existing cancer drugs. Biomark Res. 3, 9 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Watkins, J. A., Irshad, S., Grigoriadis, A. & Tutt, A. N. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 16, 211 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Lee, J. A., Carvalho, C. M. & Lupski, J. R. A DNA replication mechanism for generating nonrecurrent rearrangements associated with genomic disorders. Cell 131, 1235–1247 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Peng, J. C. & Karpen, G. H. Epigenetic regulation of heterochromatic DNA stability. Curr. Opin. Genet Dev. 18, 204–211 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Cao, L. et al. A selective requirement for 53BP1 in the biological response to genomic instability induced by Brca1 deficiency. Mol. Cell 35, 534–541 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Yazinski, S. A. et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Gene Dev. 31, 318–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Guelen, L. et al. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature 453, 948–951 (2008).

    Article  CAS  PubMed  Google Scholar 

  73. Chiolo, I. et al. Double-strand breaks in heterochromatin move outside of a dynamic HP1a domain to complete recombinational repair. Cell 144, 732–744 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ryu, T. et al. Heterochromatic breaks move to the nuclear periphery to continue recombinational repair. Nat. Cell Biol. 17, 1401–1411 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Montague, T. G., Cruz, J. M., Gagnon, J. A., Church, G. M. & Valen, E. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 42, W401–W407 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kim, S., Kim, D., Cho, S. W., Kim, J. & Kim, J. S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012–1019 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Candiano, G. et al. Blue silver: a very sensitive colloidal Coomassie G-250 staining for proteome analysis. Electrophoresis 25, 1327–1333 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Chang, H. H. Y., Watanabe, G. & Lieber, M. R. Unifying the DNA end-processing roles of the artemis nuclease. J. Biol. Chem. 290, 24036–24050 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Meister, P., Gehlen, L.R., Varela, E., Kalck, V. & Gasser, S.M. Visualizing yeast chromosomes and nuclear architecture. Methods Enzymol. 470, 535–567 (2010).

  80. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    Article  CAS  PubMed  Google Scholar 

  81. Schwanhausser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    Article  PubMed  Google Scholar 

  82. Goldman, M. J. et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 38, 675–678 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Domcke, S., Sinha, R., Levine, D. A., Sander, C. & Schultz, N. Evaluating cell lines as tumour models by comparison of genomic profiles. Nat. Commun. 4, 2126 (2013).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Huang for providing the DR-GFP and EJ5-GFP U2OS cell lines. We also thank H. Hu for providing SUM149 cells. Our work was supported by grants from the National Key Research and Development Program of China (grant no. 2018YFA0107003), the National Natural Science Foundation of China (grant nos 31930058, 32170757, 31871479 and 92249304) and the Guangdong Basic and Applied Basic Research Foundation (grant no. 2020B1515020044).

Author information

Authors and Affiliations

Authors

Contributions

B.C. and Z.S. conceptualized, designed and oversaw the project. B.C. performed the CRISPR–Cas9 screen. B.C. and T.G. performed all cell biology and microscopy experiments, with help from M.J., C.H., Y.A., S.Y., M.Y. and Y.L. B.C., L.C. and Z.H. performed protein purification and in vitro nucleolytic activity assays. R.L. and F.L. conducted the BioID experiments. Z.F., Y.X. and J.Z. conducted the bioinformatic analysis. B.C. and Z.S. wrote the manuscript, and W.M. helped with proofreading and editing.

Corresponding author

Correspondence to Zhou Songyang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks the anonymous reviewers 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 Identification of PARPi resistance genes through a genome-wide CRISPR–Cas9 screen.

a, iCas9 MDA-MB-231 cells were cultured in doxycycline (1 μg ml−1) for 3 d before single clone isolation. Individual clones were harvested for immunoblotting using an anti-Flag to detect Flag-tagged Cas9. Uninduced cells were used as a control. GAPDH served as the loading control. Clone 1 was chosen for subsequent experiments. b, iCas9 MDA-MB-231 cells (clone 1) were transduced with lentiviruses encoding paired BRCA1-specific gRNAs and then induced with doxycycline (1 μg ml−1) for 7 d before single clone isolation. Immunofluorescence analysis (left) and western blotting (right) of parental (iCas9 MDA-MB-231) and KO clone 7 (iCas9 MDA-MB-231 BRCA1−/−) cells with anti-BRCA1 are shown here. Clone 7 was used for subsequent experiments. Scale bar, 20 μm. c, Parental (WT) and BRCA1-KO (BRCA1−/−) iCas9 MDA-MB-231 cells were cultured in olaparib for 14 d at the indicated concentrations before being examined in clonogenic survival assays. d, BRCA1−/− iCas9 MDA-MB-231 cells stably expressing control (sgVector) or sgRNAs targeting REV7 or 53BP1 were cultured in doxycycline (1 μg ml−1) for 7 d. The cells were then treated with olaparib for 14 d at the indicated concentrations before clonogenic survival assays. e, Density plots showing the distribution of sgRNAs with (Day 30) and without (Day 0) olaparib treatment from the inducible CRISPR screen. f, A sample list of DDR factors and PARPi-resistance genes among high-confidence hits. Interactions from BioGRID are shown on the right. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 2 Loss of NUMEN results in PARPi resistance and sensitivity to different DNA lesions.

a, Phylogenetic tree derived from nucleotide sequences encoding NUMEN in metazoan. Phylogeny was inferred using the randomized axelerated maximum likelihood (RAxML) method with the GTRGAMMA model. Circle sizes correspond to bootstrap values from 500 replications. Evolutionary analyses were conducted in MegAlign and the tree was edited with iTOL. The scale bar represents the number of nucleotide substitutions per site. b, BRCA1−/− iCas9 MDA-MB-231 cells (Parental) were used to KO NUMEN (NUMEN−/−). GFP-tagged NUMEN (GFP–NUMEN) was then stably expressed in NUMEN and BRCA1-DKO cells. The expression of endogenous and exogenous NUMEN was confirmed by western blotting as indicated. c, A BRCA1-KO (BRCA1−/−) iCas9 HeLa cell line was similarly isolated as described above, and examined by immunofluorescence (left) and western blotting (right) for BRCA1 expression. Parental iCas9 HeLa cells served as controls. Scale bar, 20 μm. d,e, The BRCA1−/− HeLa cells from above (Parental) were used to KO NUMEN. GFP–NUMEN was then expressed in these NUMEN and BRCA1-DKO cells. NUMEN KO and rescue expression was confirmed by western blotting as indicated (d). The cells were then treated with the indicated concentrations of olaparib for 14 d before clonogenic survival assays (e). f, The BRCA1-mutant cell lines SUM149, MDA-MB-436 and HCC1937 were knocked out for NUMEN and validated by western blotting as indicated. GAPDH was used as a loading control. g,h, Parental (WT) and NUMEN-KO U2OS (g) and HeLa (h) cells were western blotted as indicated. GAPDH was used as a loading control. ik, WT and NUMEN−/− HeLa cells were subjected to the indicated dosages of IR (i), zeocin (j) and cisplatin (k) treatments before survival assessment. The cells were assessed 7 d after the treatments. Data are the mean ± s.d. of three independent experiments. Statistical analysis was performed using a two-way ANOVA. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 3 NUMEN functions as a structure-specific endo/exonuclease through its catalytic NUC domain.

a, Human NUMEN contains a putative signal peptide (SP) within the leader sequence, a conserved DNA/RNA endonuclease domain (NUC), and a C-terminus with multiple transmembrane regions (TMs). EXOG and ENDOG from the DNA/RNA non-specific endonuclease family are also shown. The RGQ/RGH motifs are marked in red. MLS indicates mitochondrial localization sequence. b,c, GFP-tagged full-length NUMEN (+GFP–NUMEN) or the NUC-deletion mutant (+GFP–NUMEN-ΔNUC) was stably expressed in BRCA1 and NUMEN-DKO (BRCA1−/−NUMEN−/−) MDA-MB-231 cells. b, Exogenous NUMEN expression was confirmed by western blotting using anti-GFP or anti-α-tubulin. c, The cells were cultured for 14 d in olaparib as indicated before clonogenic survival assays. Parental BRCA1−/− MDA-MB-231 cells served as controls. d, Recombinant full-length and mutant NUMEN NUC domain proteins were purified from E. coli using nickel-agarose columns and analysed by SDS–PAGE and Coomassie blue staining. e, 32P-labelled DNA substrates (40 nM) were incubated with (+) or without (−) NUMEN NUC proteins (50 nM) for 50 min at 37 °C. Stars indicate 5′-end labelling with 32P. The red arrowheads indicate how NUMEN processes the substrates from blunt-ended termini and/or past ds/ss junctions. The red arrows indicate the direction of processing. f, Alignment of the NUC domain sequences of NUMEN, ENDOG and EXOG from human and other species. The secondary structure of human NUMEN was obtained from AlphaFold 2. Multiple sequence alignments were performed using CLUSTALW and displayed by ESPript 3.0. The red and green stars underneath the sequences denote residue Gln143 and evolutionarily conserved cysteine residues, respectively. The β strands are marked by arrows and helices by spirals. Identical amino acids are shaded in red and conserved residues are boxed. g, ss- and dsDNA substrates were incubated with 50 nM recombinant WT or RGQ point mutants of NUMEN NUC domains for 40 min at 37 °C. Reactions with no NUC proteins (Mock) served as controls. Cy5 labelling is indicated by asterisks and biotin-TEG modifications by ellipses. h, GFP–NUMEN was stably expressed in MDA-MB-231 cells. Endogenous and exogenous NUMEN expression in WT, NUMEN−/− and NUMEN overexpression (GFP–NUMEN) cells was confirmed by western blotting as indicated. Source unprocessed blots are provided.

Source data

Extended Data Fig. 4 NUMEN deficiency impairs NHEJ and promotes HR.

a, HA-tagged ER-AsiSI was stably expressed in U2OS cells and the the indicated antibodies were used in western blots. b, Cells from a were cultured in 300 nM 4-OHT for 4 h and then immunostained using antibodies to the HA epitope (red). Induced translocation of ER-AsisI into the nucleus could be observed. DSBs generated by the restriction enzyme AsisI were visualized using anti-γH2AX (green). Representative images from confocal microscopy are shown. DNA was counterstained with DAPI (blue). Scale bars, 5 μm. c) DR-GFP and EJ5-GFP U2OS cell lines were transiently transfected with siRNAs targeting NUMEN. The knockdown efficiency was confirmed by western blotting, as indicated. d, Gating strategy used for Fig. 3d. The first gating was performed using FSC-A and SSC-A, the second gating was performed using FITC-A and PE-A. e, WT and NUMEN−/− MDA-MB-231 cells were treated with or without IR (2 Gy) and harvested 2 h later for immunostaining experiments using anti-RAD51. Data are the mean ± s.d. of three independent experiments. Circles are the mean values from each experiment; 30 nuclei were analysed in each group per experiment. Significance was calculated using a two-way ANOVA. NS, non-significant. Scale bar, 5 μm. f, Representative images of immunostaining analysis of WT and NUMEN−/− MDA-MB-231 cells that were harvested at the indicated time points after zeocin addition (500 μg ml−1), using antibodies to 53BP1 (left) or γH2AX (right). The cells were maintained without zeocin after 4 h. Scale bar, 5 μm. g, WT, TRF2−/− and TRF2 and NUMEN-DKO (NUMEN−/−TRF2−/−) HeLa cells were immunoblotted using anti-TRF2 and anti-GAPDH. h, BRCA1−/− and BRCA1 and NUMEN-DKO (BRCA1−/−NUMEN−/−) HeLa cells were treated with DMSO or zeocin (100 μg ml−1) for 4 h before being analysed for RAD51 foci formation. Data represent the mean ± s.d. of three independent experiments (n = 50 nuclei per condition per experiment). Significance was calculated using a two-way ANOVA. NS, non-significant. Scale bar, 5 μm. Source numerical data and unprocessed blots are provided.

Source data

Extended Data Fig. 5 Identification of the interaction network of NUMEN through BioID proximity labelling strategy.

a,b, BioID proximity screen. a, HEK293T cells were transduced with lentiviruses encoding Flag–BioID-tagged NUMEN and cultured in biotin-containing media before being harvested for large-scale affinity purification and identification of biotinylated proteins by mass spectrometry. Flag–BioID fused to an NLS served as the control. Two repeat control experiments were performed. b, Flag–BioID was inserted internally either N-terminal (BioID-NUMEN) or C-terminal (NUMEN–BioID) of the NUC domain. c, Enrichment patterns of the proteins identified from the two control repeat experiments (Ctrl-1 and Ctrl-2) versus the two NUMEN baits. d, Number of overlapping candidate proteins between different sample groups. e, The degree of correlation between control repeats and the two NUMEN baits is shown using log2-transformed FOT values. f, Gene Ontology enrichment analysis of cellular component for the preys identified from both NUMEN baits. The ER and nuclear membrane-associated terms are marked in blue and red, respectively. The top 20 enriched Gene Ontology terms under the ‘cellular component’ category for the baits are listed. Orange and grey bars represent respectively fold enrichments and −log10-transformed P values. g, Nuclear membrane proteins identified from our BioID screens are plotted as shown. The nuclear envelope LINC complex and LAD proteins are illustrated (top). The top-right quadrant of NUMEN–BioID scatter plot is shown (bottom), with enriched proteins (log2-transformed ratio) over controls. Significantly enriched nuclear membrane proteins (LINC complex in orange and LAD proteins in blue) and DDR factors (red) are highlighted. Putative NUMEN-interacting partners based on the BioGRID database are marked in black. Dashed lines indicate 1.5-fold change of FOT values.

Extended Data Fig. 6 Epistasis analysis of NUMEN on the NHEJ repair pathway.

a, siRNAs taregting the indicated genes were transiently transfected into U2OS EJ5-GFP and HeLa cells. The knockdown efficiencies were determined by RT–qPCR. A non-silencing siRNA (siNC) served as the control. Data are the mean ± s.d. of three independent experiments. Significance was calculated using a two-tailed unpaired t-test. b, siRNAs targeting the indicated gene products were introduced into U2OS EJ5-GFP cells before the NHEJ efficiencies were measured. All results were normalized to the control sample. Data are the mean ± s.d. of three different experiments. Statistical analysis was performed using a one-way ANOVA. NS, not significant. c, WT and NUMEN−/− MDA-MB-231 cells were treated with the DNA-PK inhibitor NU7441 (1 μM for 24 h) or knocked out for Artemis. The cells were then treated with zeocin (100 μg ml−1) for 4 h before immunostaining analysis using anti-RAD51. Data are the mean ± s.d. (n = 3 independent experiments). Quantification was carried out on 30 nuclei in each group per experiment. Statistical analysis was performed using a one-way ANOVA. NS, not significant. d, WT and NUMEN−/− MDA-MB-231 cells were treated with DMSO or 100 µg ml−1 zeocin for 4 h before immunofluorescence analysis and quantification. An antibody to phospho-DNA-PK S2056 (p-DNA-PK) was used. Data are the mean ± s.d. of three independent experiments (n = 30 nuclei per condition per experiment). Statistical analysis was performed using a one-way ANOVA. NS, not significant. e, 53BP1 was knocked down in WT and NUMEN−/− HeLa cells before treatment with the indicated concentrations of zeocin for 4 h. The cells were then maintained in normal culture medium for 7 d and then assessed for survival. f, WT and NUMEN−/− HeLa cells were knocked out for 53BP1 and subjected to zeocin (100 μg ml−1) treatment for 4 h before immunostaining using anti-RAD51. Data represent the mean ± s.d. (n = 3 independent experiments, 30 nuclei were analysed per experiment). Significance was calculated using two-tailed unpaired t-tests. NS, not significant. Source numerical data are provided.

Source data

Extended Data Fig. 7 NUMEN is anchored to the nuclear membrane through its SP and TM domains.

a, MDA-MB-231, HeLa and HEK293T cells were co-stained with antibodies to NUMEN (green) and Lamin A/C (red). Representative images from confocal microscopy are shown. DNA was counterstained with DAPI (blue). b, HEK293T cells transiently expressing Flag-tagged full-length or truncation mutants of NUMEN were co-stained with antibodies to Lamin A/C (red) and Flag (green). Representative images from confocal microscopy are shown. The Flag epitope was inserted C-terminal of the SP sequence whenever possible to avoid disrupting NUMEN localization. c, Representative images of HEK293T cells stably expressing BioID-Flag-tagged NUMEN that were immunostained with antibodies to Flag (green) and Lamin A/C (red). d, Different NUMEN mutants were stably expressed in MDA-MB-231 cells and their expression confirmed by western blotting with antibodies to the Flag epitope. GAPDH was used as a loading control. e, Cells from d were treated with zeocin (100 μg ml−1) for 4 h before immunostaining analysis using anti-RAD51. Data represent the mean ± s.d. (n = 3 independent experiments). Quantification was carried out on 30 nuclei in each group per experiment. Significance was calculated using two-tailed unpaired t-tests. ad, Scale bars, 5 μm. Source numerical data are provided.

Source data

Extended Data Fig. 8 NUMEN participates in compartmentalized NHEJ repair at the nuclear periphery.

a, HEK293T cells stably expressing the 53BP1trunc–mApple DSB reporter (red) together with NUMEN–GFP (green) were cultured with zeocin (100 μg ml−1) in a glass-bottomed dish before time-lapse live-cell imaging. 53BP1trunc–mApple is a fusion protein of truncated 53BP1 and the Apple fluorescent protein. Images were captured every 3 min during a 4-h window, and those from the indicated time points are shown. Each boxed region is enlarged in the zoom panel (bottom). Arrows point to 53BP1trunc–mApple signals that moved closer to NUMEN–GFP signals over time. b, The 53BP1trunc–mApple DSB reporter cells described above were treated with 2 Gy IR and visualized by time-lapse live-cell imaging. Images were captured every 3 min during a 2-h window. c, GFP-tagged m6A-Tracer and Dam-LaminB1–mAmetrine co-expression HEK293T cells were cultured with (+) or without (−) the Shield-1 ligand for 24 h. In the absence of Shield-1 ligand, only m6A-Tracer (green) was expressed and showed predominantly cytoplasmic localization. Following induction, Dam-Lamin B1 (blue) became stabilized and along with m6A exhibited ring-like localization patterns at the nuclear lamina. d, HEK293T cells stably co-expressing m6A-Tracer–GFP (green, to mark LADs) and Dam-Lamin B1–mAmetrine were cultured with the Shield-1 ligand and immunostained using anti-γH2AX (red) following 100 μg ml−1 zeocin treatment for 4 h. The boxed region is enlarged in the zoom panel. Scale bars, 5 μm, unless specified otherwise in the zoom panel. Source numerical data are provided.

Extended Data Fig. 9 NUMEN enables in situ NHEJ repair of heterochromatic breaks and maintaining of genome stability.

a, MDA-MB-231 cells were treated with DMSO, zeocin (100 μg ml−1 for 4 h), bleomycin (40 μg ml−1 for 4 h), IR (2 Gy and harvested 2 h later) or cisplatin (2.5 μM for 4 h) before immunofluorescence analysis using antibodies to γH2AX (green) and H3K9me2 (red). DAPI was used to stain the nuclei (blue). Scale bars, 5 μm. b, Analysis of CNV counts from four cancer types with significant negative correlations from a. LIHC (n = 337), COAD (n = 407), HNSC (n = 468), and BRCA (n = 1,027) samples with valid outcome data were grouped by the expression levels of BRCA1 and NUMEN (median cutoff) and are shown in violin plots. Each box outline shows the 25th and 75th percentiles, and the solid line indicates the median value. Whiskers extend to the most extreme data points that are no more than 1.5× the interquartile range. n, number of subgroup samples. Statistical analysis was performed using a two-tailed Mann–Whitney U-test. c, DSB repair pathway choice is tightly regulated to ensure the faithful repair of chromosome breaks. Nuclear compartmentation may help shape such decisions. We propose that nuclear membrane anchoring of NUMEN facilitates the establishment of an environment that favours NHEJ for the repair of repetitive DNA sequences at LADs as well as lesions dynamically moving from the nuclear interior to the periphery. NUMEN may thus antagonize HDR near the nuclear periphery and help minimize ectopic repair between heterochromatic repetitive sequences. Source numerical data are provided.

Supplementary information

Reporting Summary

Supplementary Table 1

Complete list of MAGeCK analysis of PARPi resistance CRISPR screen.

Supplementary Table 2

The proximity/interaction proteins of NUMEN identified by mass spectrometry from the BioID pulldown expriments.

Supplementary Tables 3 and 4

Supplementary Table 3. List of oligonucleotides used in the study. Supplementary Table 4. List of antibodies used in the study.

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

Chen, B., Ge, T., Jian, M. et al. Transmembrane nuclease NUMEN/ENDOD1 regulates DNA repair pathway choice at the nuclear periphery. Nat Cell Biol 25, 1004–1016 (2023). https://doi.org/10.1038/s41556-023-01165-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01165-1

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