Abstract
Bursts of cell proliferation after infection, injury or transformation can coincide with DNA damage and spindle assembly defects. These increase the risk of cell cycle arrest in mitosis, during which many cellular processes are uniquely regulated. Ultimately, cells arrested during mitosis may die, but adaptive mechanisms also allow their escape into the next interphase. This step can have variable consequences, including chromosome missegregation, polyploidization and centrosome amplification. Escaping cells can also initiate innate immune signalling, enter senescence or engage cell death, which in turn alert the microenvironment through nucleic acid sensing mechanisms and/or the release of danger-associated molecular patterns. Here we discuss the causes and consequences of deregulated mitosis and postmitotic cell fate, highlighting the impact of DNA damage repair, the spindle assembly checkpoint and extra centrosomes on genome integrity, as well as inflammatory signalling. Finally, we attempt to reconcile conflicting observations and propose variable modes that activate innate immune responses after mitotic perturbations.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
References
Matthews, H. K., Bertoli, C. & de Bruin, R. A. M. Cell cycle control in cancer. Nat. Rev. Mol. Cell Biol. 23, 74–88 (2022).
Costa, A. & Diffley, J. F. X. The initiation of eukaryotic DNA replication. Annu. Rev. Biochem. 91, 107–131 (2022).
Davidson, I. F. & Peters, J. M. Genome folding through loop extrusion by SMC complexes. Nat. Rev. Mol. Cell Biol. 22, 445–464 (2021).
Breslow, D. K. & Holland, A. J. Mechanism and regulation of centriole and cilium biogenesis. Annu. Rev. Biochem. 88, 691–724 (2019).
Dey, G. & Baum, B. Nuclear envelope remodelling during mitosis. Curr. Opin. Cell Biol. 70, 67–74 (2021).
Gibcus, J.H, et al. A pathway for mitotic chromosome formation. Science 359, eaao6135 (2018).
Zhao, G. et al. A tubule-sheet continuum model for the mechanism of nuclear envelope assembly. Dev. Cell 58, 847–865 (2023).
Palozola, K. C. et al. Mitotic transcription and waves of gene reactivation during mitotic exit. Science 358, 119–122 (2017).
Tanenbaum, M. E., Stern-Ginossar, N., Weissman, J. S. & Vale, R. D. Regulation of mRNA translation during mitosis. eLife 4, e07957 (2015).
Ly, J. et al. Nuclear release of eIF1 restricts start-codon selection during mitosis. Nature 635, 490–498 (2024).
Valdez, V. A., Neahring, L., Petry, S. & Dumont, S. Mechanisms underlying spindle assembly and robustness. Nat. Rev. Mol. Cell Biol. 24, 523–542 (2023).
Salinas-Luypaert, C. & Fachinetti, D. Canonical and noncanonical regulators of centromere assembly and maintenance. Curr. Opin. Cell Biol. 89, 102396 (2024).
Waterman, D. P., Haber, J. E. & Smolka, M. B. Checkpoint responses to DNA double-strand breaks. Annu. Rev. Biochem. 89, 103–133 (2020).
Maiato, H. & Silva, S. Double-checking chromosome segregation. J. Cell Biol. 222, e202301106 (2023).
McAinsh, A. D. & Kops, G. Principles and dynamics of spindle assembly checkpoint signalling. Nat. Rev. Mol. Cell Biol. 24, 543–559 (2023).
Ganem, N. J., Godinho, S. A. & Pellman, D. A mechanism linking extra centrosomes to chromosomal instability. Nature 460, 278–282 (2009).
Dwivedi, D., Harry, D. & Meraldi, P. Mild replication stress causes premature centriole disengagement via a sub-critical Plk1 activity under the control of ATR−Chk1. Nat. Commun. 14, 6088 (2023).
Haschka, M., Karbon, G., Fava, L. L. & Villunger, A. Perturbing mitosis for anti-cancer therapy: is cell death the only answer? EMBO Rep. 19, e45440 (2018).
Szmyd, R. et al. Homologous recombination promotes non-immunogenic mitotic cell death upon DNA damage. Nat. Cell Biol. 27, 59–72 (2025).
Flynn, F., Koch, P. D. & Mitchison, T. J. Chromatin bridges, not micronuclei, activate cGAS after drug-induced mitotic errors in human cells. Proc. Natl Acad. Sci. USA 118, e2103585118 (2021).
Masamsetti, V. P. et al. Replication stress induces mitotic death through parallel pathways regulated by WAPL and telomere deprotection. Nat. Commun. 10, 4224 (2019).
Garner, E., Kim, Y., Lach, F. P., Kottemann, M. C. & Smogorzewska, A. Human GEN1 and the SLX4-associated nucleases MUS81 and SLX1 are essential for the resolution of replication-induced Holliday junctions. Cell Rep. 5, 207–215 (2013).
Chen, J. et al. Cell cycle checkpoints cooperate to suppress DNA- and RNA-associated molecular pattern recognition and anti-tumor immune responses. Cell Rep. 32, 108080 (2020).
Hayashi, M. T., Cesare, A. J., Fitzpatrick, J. A., Lazzerini-Denchi, E. & Karlseder, J. A telomere-dependent DNA damage checkpoint induced by prolonged mitotic arrest. Nat. Struct. Mol. Biol. 19, 387–394 (2012).
Lemmens, B. et al. DNA replication determines timing of mitosis by restricting CDK1 and PLK1 activation. Mol. Cell 71, 117–128 (2018).
Saldivar, J. C. et al. An intrinsic S/G(2) checkpoint enforced by ATR. Science 361, 806–810 (2018).
Minocherhomji, S. et al. Replication stress activates DNA repair synthesis in mitosis. Nature 528, 286–290 (2015).
Groelly, F. J. et al. Mitotic DNA synthesis is caused by transcription-replication conflicts in BRCA2-deficient cells. Mol. Cell 82, 3382–3397 (2022).
Lukas, C. et al. 53BP1 nuclear bodies form around DNA lesions generated by mitotic transmission of chromosomes under replication stress. Nat. Cell Biol. 13, 243–253 (2011).
Gelot, C. et al. Polθ is phosphorylated by PLK1 to repair double-strand breaks in mitosis. Nature 621, 415–422 (2023).
Mazzagatti, A., Engel, J. L. & Ly, P. Boveri and beyond: chromothripsis and genomic instability from mitotic errors. Mol. Cell 84, 55–69 (2024).
Hatch, E. M., Fischer, A. H., Deerinck, T. J. & Hetzer, M. W. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 154, 47–60 (2013).
Crasta, K. et al. DNA breaks and chromosome pulverization from errors in mitosis. Nature 482, 53–58 (2012).
Zhang, C. Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 179–184 (2015).
Umbreit, N. T. et al. Mechanisms generating cancer genome complexity from a single cell division error. Science 368, eaba0712 (2020).
Engel, J. L. et al. The Fanconi anemia pathway induces chromothripsis and ecDNA-driven cancer drug resistance. Cell 187, 6055–6070 (2024).
Topham, C. et al. MYC is a major determinant of mitotic cell fate. Cancer Cell 28, 129–140 (2015).
Haschka, M. D. et al. The NOXA–MCL1–BIM axis defines lifespan on extended mitotic arrest. Nat. Commun. 6, 6891 (2015).
Topham, C. H. & Taylor, S. S. Mitosis and apoptosis: how is the balance set? Curr. Opin. Cell Biol. 25, 780–785 (2013).
Díaz-Martínez, L. A. et al. Genome-wide siRNA screen reveals coupling between mitotic apoptosis and adaptation. EMBO J. 33, 1960–1976 (2014).
Lok, T. M. et al. Mitotic slippage is determined by p31comet and the weakening of the spindle-assembly checkpoint. Oncogene 39, 2819–2834 (2020).
Tuzlak, S., Kaufmann, T. & Villunger, A. Interrogating the relevance of mitochondrial apoptosis for vertebrate development and postnatal tissue homeostasis. Genes Dev. 30, 2133–2151 (2016).
Czabotar, P. E. & Garcia-Saez, A. J. Mechanisms of BCL-2 family proteins in mitochondrial apoptosis. Nat. Rev. Mol. Cell Biol. 24, 732–748 (2023).
Glover, H. L., Schreiner, A., Dewson, G. & Tait, S. W. G. Mitochondria and cell death. Nat. Cell Biol. 26, 1434–1446 (2024).
Shi, J. & Mitchison, T. J. Cell death response to anti-mitotic drug treatment in cell culture, mouse tumor model and the clinic. Endocr. Relat. Cancer 24, T83–T96 (2017).
Bock, F. J. & Riley, J. S. When cell death goes wrong: inflammatory outcomes of failed apoptosis and mitotic cell death. Cell Death Differ. 30, 293–303 (2023).
Darweesh, O. et al. Identification of a novel Bax-Cdk1 signalling complex that links activation of the mitotic checkpoint to apoptosis. J. Cell Sci. 134, jcs244152 (2021).
Allan, L. A., Skowyra, A., Rogers, K. I., Zeller, D. & Clarke, P. R. Atypical APC/C-dependent degradation of Mcl-1 provides an apoptotic timer during mitotic arrest. EMBO J. 37, e96831 (2018).
Arai, S. et al. MARCH5 mediates NOXA-dependent MCL1 degradation driven by kinase inhibitors and integrated stress response activation. eLife 9, e54954 (2020).
Haschka, M. D. et al. MARCH5-dependent degradation of MCL1/NOXA complexes defines susceptibility to antimitotic drug treatment. Cell Death Differ. 27, 2297–2312 (2020).
Wertz, I. E. et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature 471, 110–114 (2011).
Shi, J., Zhou, Y., Huang, H. C. & Mitchison, T. J. Navitoclax (ABT-263) accelerates apoptosis during drug-induced mitotic arrest by antagonizing Bcl-xL. Cancer Res. 71, 4518–4526 (2011).
Nikhil, K. & Shah, K. The Cdk5–Mcl-1 axis promotes mitochondrial dysfunction and neurodegeneration in a model of Alzheimer’s disease. J. Cell Sci. 130, 3023–3039 (2017).
Lowman, X. H. et al. The proapoptotic function of Noxa in human leukemia cells is regulated by the kinase Cdk5 and by glucose. Mol. Cell 40, 823–833 (2010).
Zheng, X. F. et al. CDK5–cyclin B1 regulates mitotic fidelity. Nature 633, 932–940 (2024).
Zierhut, C. et al. The cytoplasmic DNA sensor cGAS promotes mitotic cell death. Cell 178, 302–315 (2019).
Moustafa-Kamal, M., Gamache, I., Lu, Y., Li, S. & Teodoro, J. G. BimEL is phosphorylated at mitosis by Aurora A and targeted for degradation by βTrCP1. Cell Death Differ. 20, 1393–1403 (2013).
Tanaka, K. et al. Targeting Aurora B kinase prevents and overcomes resistance to EGFR inhibitors in lung cancer by enhancing BIM- and PUMA-mediated apoptosis. Cancer Cell 39, 1245–1261 (2021).
Vaz, S. et al. FOXM1 repression increases mitotic death upon antimitotic chemotherapy through BMF upregulation. Cell Death Dis. 12, 542 (2021).
Pedley, R. et al. BioID-based proteomic analysis of the Bid interactome identifies novel proteins involved in cell-cycle-dependent apoptotic priming. Cell Death Dis. 11, 872 (2020).
Karbon, G. et al. The BH3-only protein NOXA serves as an independent predictor of breast cancer patient survival and defines susceptibility to microtubule targeting agents. Cell Death Dis. 12, 1151 (2021).
Tan, T. T. et al. Key roles of BIM-driven apoptosis in epithelial tumors and rational chemotherapy. Cancer Cell 7, 227–238 (2005).
Karbon, G. et al. Chronic spindle assembly checkpoint activation causes myelosuppression and gastrointestinal atrophy. EMBO Rep. 25, 2743–2772 (2024).
Matthess, Y., Raab, M., Knecht, R., Becker, S. & Strebhardt, K. Sequential Cdk1 and Plk1 phosphorylation of caspase-8 triggers apoptotic cell death during mitosis. Mol. Oncol. 8, 596–608 (2014).
Andersen, J. L. et al. Restraint of apoptosis during mitosis through interdomain phosphorylation of caspase-2. EMBO J. 28, 3216–3227 (2009).
Connolly, P., Garcia-Carpio, I. & Villunger, A. Cell-cycle cross talk with caspases and their substrates. Cold Spring Harb. Perspect. Biol. 12, a036475 (2020).
Allan, L. A. & Clarke, P. R. Phosphorylation of caspase-9 by CDK1/cyclin B1 protects mitotic cells against apoptosis. Mol. Cell 26, 301–310 (2007).
Lim, Y. et al. Phosphorylation by Aurora B kinase regulates caspase-2 activity and function. Cell Death Differ. 28, 349–366 (2021).
Van Opdenbosch, N. & Lamkanfi, M. Caspases in cell death, inflammation, and disease. Immunity 50, 1352–1364 (2019).
Moore, A. S. et al. Actin cables and comet tails organize mitochondrial networks in mitosis. Nature 591, 659–664 (2021).
Meier, P., Legrand, A. J., Adam, D. & Silke, J. Immunogenic cell death in cancer: targeting necroptosis to induce antitumour immunity. Nat. Rev. Cancer 24, 299–315 (2024).
Liccardi, G. et al. RIPK1 and caspase-8 ensure chromosome stability independently of their role in cell death and inflammation. Mol. Cell 73, 413–428 (2019).
Pollak, N. et al. Cell cycle progression and transmitotic apoptosis resistance promote escape from extrinsic apoptosis. J. Cell Sci. 134, jcs258966 (2021).
Brito, D. A. & Rieder, C. L. Mitotic checkpoint slippage in humans occurs via cyclin B destruction in the presence of an active checkpoint. Curr. Biol. 16, 1194–1200 (2006).
Meitinger, F. et al. 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. J. Cell Biol. 214, 155–166 (2016).
Lambrus, B. G. et al. A USP28–53BP1–p53–p21 signaling axis arrests growth after centrosome loss or prolonged mitosis. J. Cell Biol. 214, 143–153 (2016).
Fong, C. S. et al. 53BP1 and USP28 mediate p53-dependent cell cycle arrest in response to centrosome loss and prolonged mitosis. eLife 5, e16270 (2016).
Uetake, Y. & Sluder, G. Prolonged prometaphase blocks daughter cell proliferation despite normal completion of mitosis. Curr. Biol. 20, 1666–1671 (2010).
Meitinger, F. et al. Control of cell proliferation by memories of mitosis. Science 383, 1441–1448 (2024).
Burigotto, M. et al. PLK1 promotes the mitotic surveillance pathway by controlling cytosolic 53BP1 availability. EMBO Rep. 24, e57234 (2023).
Bazzi, H. & Anderson, K. V. Acentriolar mitosis activates a p53-dependent apoptosis pathway in the mouse embryo. Proc. Natl Acad. Sci. USA 111, E1491–E1500 (2014).
Xiao, C. et al. Gradual centriole maturation associates with the mitotic surveillance pathway in mouse development. EMBO Rep. 22, e51127 (2021).
Meyer-Gerards, C. & Bazzi, H. Developmental and tissue-specific roles of mammalian centrosomes. FEBS J. 292, 709–726 (2025).
Phan, T. P. & Holland, A. J. Time is of the essence: the molecular mechanisms of primary microcephaly. Genes Dev. 35, 1551–1578 (2021).
Schapfl, M. A. et al. Centrioles are frequently amplified in early B cell development but dispensable for humoral immunity. Nat. Commun. 15, 8890 (2024).
Collin, P., Nashchekina, O., Walker, R. & Pines, J. The spindle assembly checkpoint works like a rheostat rather than a toggle switch. Nat. Cell Biol. 15, 1378–1385 (2013).
Fulcher, L. J., Sobajima, T., Batley, C., Gibbs-Seymour, I. & Barr, F. A. MDM2 functions as a timer reporting the length of mitosis. Nat. Cell Biol. 27, 262–272 (2025).
Herve, S. et al. Chromosome mis-segregation triggers cell cycle arrest through a mechanosensitive nuclear envelope checkpoint. Nat. Cell Biol. 27, 73–86 (2025).
Chunduri, N. K. & Storchová, Z. The diverse consequences of aneuploidy. Nat. Cell Biol. 21, 54–62 (2019).
Ben-David, U. & Amon, A. Context is everything: aneuploidy in cancer. Nat. Rev. Genet. 21, 44–62 (2020).
Ippolito, M. R. et al. Increased RNA and protein degradation is required for counteracting transcriptional burden and proteotoxic stress in human aneuploid cells. Cancer Discov. 14, 2532–2553 (2024).
Ottens, F., Efstathiou, S. & Hoppe, T. Cutting through the stress: RNA decay pathways at the endoplasmic reticulum. Trends Cell Biol. 34, 1056–1068 (2024).
Xu, L. et al. IRE1α silences dsRNA to prevent taxane-induced pyroptosis in triple-negative breast cancer. Cell 187, 7248–7266 (2024).
Santaguida, S., Vasile, E., White, E. & Amon, A. Aneuploidy-induced cellular stresses limit autophagic degradation. Genes Dev. 29, 2010–2021 (2015).
Stingele, S. et al. Global analysis of genome, transcriptome and proteome reveals the response to aneuploidy in human cells. Mol. Syst. Biol. 8, 608 (2012).
Zerbib, J. et al. Human aneuploid cells depend on the RAF/MEK/ERK pathway for overcoming increased DNA damage. Nat. Commun. 15, 7772 (2024).
Ippolito, M. R. et al. Gene copy-number changes and chromosomal instability induced by aneuploidy confer resistance to chemotherapy. Dev. Cell 56, 2440–2454 (2021).
Cohen-Sharir, Y. et al. Aneuploidy renders cancer cells vulnerable to mitotic checkpoint inhibition. Nature 590, 486–491 (2021).
Zheng, S. et al. High CDC20 levels increase sensitivity of cancer cells to MPS1 inhibitors. EMBO Rep. 26, 1036–1061 (2025).
Bennett, A. et al. Inhibition of Bcl-xL sensitizes cells to mitotic blockers, but not mitotic drivers. Open Biol. 6, 160134 (2016).
Weiss, J. G., Gallob, F., Rieder, P. & Villunger, A. Apoptosis as a barrier against CIN and aneuploidy. Cancers 15, 30 (2022).
Boon, N. J. et al. DNA damage induces p53-independent apoptosis through ribosome stalling. Science 384, 785–792 (2024).
Ogawa, A. et al. SLFN11-mediated ribosome biogenesis impairment induces TP53-independent apoptosis. Mol. Cell 85, 894–912 (2025).
Piñon, J. D., Labi, V., Egle, A. & Villunger, A. Bim and Bmf in tissue homeostasis and malignant disease. Oncogene 27, S41–S52 (2008).
Fava, L. L. et al. The PIDDosome activates p53 in response to supernumerary centrosomes. Genes Dev. 31, 34–45 (2017).
Evans, L. T. et al. ANKRD26 recruits PIDD1 to centriolar distal appendages to activate the PIDDosome following centrosome amplification. EMBO J. 40, e105106 (2021).
Quintyne, N. J., Reing, J. E., Hoffelder, D. R., Gollin, S. M. & Saunders, W. S. Spindle multipolarity is prevented by centrosomal clustering. Science 307, 127–129 (2005).
Burigotto, M. et al. Centriolar distal appendages activate the centrosome–PIDDosome–p53 signalling axis via ANKRD26. EMBO J. 40, e104844 (2021).
Sladky, V. C. et al. E2F-family members engage the PIDDosome to limit hepatocyte ploidy in liver development and regeneration. Dev. Cell 52, 335–349 (2020).
Rizzotto, D., Englmaier, L. & Villunger, A. At a crossroads to cancer: how p53-induced cell fate decisions secure genome integrity. Int. J. Mol. Sci. 22, 10883 (2021).
Rizzotto, D. et al. Caspase-2 kills cells with extra centrosomes. Sci. Adv. 10, eado6607 (2024).
Edwards, F., et al. Centrosome amplification primes ovarian cancer cells for apoptosis and potentiates the response to chemotherapy. PLoS Biol. 22, e3002759 (2024).
McLaughlin, M. et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat. Rev. Cancer 20, 203–217 (2020).
Di Maggio, F. M. et al. Portrait of inflammatory response to ionizing radiation treatment. J. Inflamm.12, 14 (2015).
Mackenzie, K. J. et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465 (2017).
Zhang, B., Xu, P. & Ablasser, A. Regulation of the cGAS–STING pathway. Annu. Rev. Immunol. 43, 667–692 (2025).
Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).
Wu, J. et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339, 826–830 (2013).
Gulen, M. F. et al. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 8, 427 (2017).
Glück, S. et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat. Cell Biol. 19, 1061–1070 (2017).
Yang, H., Wang, H., Ren, J., Chen, Q. & Chen, Z. J. cGAS is essential for cellular senescence. Proc. Natl Acad. Sci. USA 114, E4612–E4620 (2017).
Bakhoum, S. F. et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 553, 467–472 (2018).
Hong, C. et al. cGAS–STING drives the IL-6-dependent survival of chromosomally instable cancers. Nature 607, 366–373 (2022).
Volkman, H. E., Cambier, S., Gray, E. E. & Stetson, D. B. Tight nuclear tethering of cGAS is essential for preventing autoreactivity. eLife 8, e47491 (2019).
Xu, P. et al. The CRL5–SPSB3 ubiquitin ligase targets nuclear cGAS for degradation. Nature 627, 873–879 (2024).
Xie, W. & Patel, D. J. Keeping innate immune response in check: when cGAS meets the nucleosome. Cell Res. 30, 1055–1056 (2020).
Harding, S. M. et al. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548, 466–470 (2017).
Dou, Z. et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550, 402–406 (2017).
Mackenzie, K. J. et al. Ribonuclease H2 mutations induce a cGAS/STING-dependent innate immune response. EMBO J. 35, 831–844 (2016).
Zierhut, C. & Funabiki, H. Regulation and consequences of cGAS activation by self-DNA. Trends Cell Biol. 30, 594–605 (2020).
Wang, B., Han, J., Elisseeff, J. H. & Demaria, M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat. Rev. Mol. Cell Biol. 25, 958–978 (2024).
De Cecco, M. et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 566, 73–78 (2019).
Victorelli, S. et al. Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature 622, 627–636 (2023).
López-Polo, V. et al. Release of mitochondrial dsRNA into the cytosol is a key driver of the inflammatory phenotype of senescent cells. Nat. Commun. 15, 7378 (2024).
Uggenti, C., Lepelley, A. & Crow, Y. J. Self-awareness: nucleic acid-driven inflammation and the type I interferonopathies. Annu. Rev. Immunol. 37, 247–267 (2019).
Krupina, K., Goginashvili, A. & Cleveland, D. W. Causes and consequences of micronuclei. Curr. Opin. Cell Biol. 70, 91–99 (2021).
Lebrec, V. et al. A microscopy reporter for cGAMP reveals rare cGAS activation following DNA damage, and a lack of correlation with micronuclear cGAS enrichment. Preprint at bioRxiv https://doi.org/10.1101/2024.05.13.593978 (2024).
Takaki, T., Millar, R., Hiley, C. T. & Boulton, S. J. Micronuclei induced by radiation, replication stress, or chromosome segregation errors do not activate cGAS–STING. Mol. Cell 84, 2203–2213 (2024).
Sato, Y. & Hayashi, M. T. Micronucleus is not a potent inducer of the cGAS/STING pathway. Life Sci. Alliance 7, e202302424 (2024).
Smarduch, S. et al. A novel biosensor for the spatiotemporal analysis of STING activation during innate immune responses to dsDNA. EMBO J. 44, 2157–2182 (2025).
Agustinus, A. S. et al. Epigenetic dysregulation from chromosomal transit in micronuclei. Nature 619, 176–183 (2023).
Coquel, F. et al. SAMHD1 acts at stalled replication forks to prevent interferon induction. Nature 557, 57–61 (2018).
Crossley, M. P. et al. R-loop-derived cytoplasmic RNA–DNA hybrids activate an immune response. Nature 613, 187–194 (2023).
Mankan, A. K. et al. Cytosolic RNA:DNA hybrids activate the cGAS–STING axis. EMBO J.33, 2937–2946 (2014).
Wang, Y. et al. eccDNAs are apoptotic products with high innate immunostimulatory activity. Nature 599, 308–314 (2021).
Yang, F. et al. Retrotransposons hijack alt-EJ for DNA replication and eccDNA biogenesis. Nature 620, 218–225 (2023).
Tang, S., Stokasimov, E., Cui, Y. & Pellman, D. Breakage of cytoplasmic chromosomes by pathological DNA base excision repair. Nature 606, 930–936 (2022).
Bhowmick, R., Hickson, I. D. & Liu, Y. Completing genome replication outside of S phase. Mol. Cell 83, 3596–3607 (2023).
Deng, L. et al. Mitotic CDK promotes replisome disassembly, fork breakage, and complex DNA rearrangements. Mol. Cell 73, 915–929 (2019).
Leimbacher, P. A. et al. MDC1 interacts with TOPBP1 to maintain chromosomal stability during mitosis. Mol. Cell 74, 571–583 (2019).
Trivedi, P., Steele, C. D., Au, F. K. C., Alexandrov, L. B. & Cleveland, D. W. Mitotic tethering enables inheritance of shattered micronuclear chromosomes. Nature 618, 1049–1056 (2023).
Lin, Y. F. et al. Mitotic clustering of pulverized chromosomes from micronuclei. Nature 618, 1041–1048 (2023).
De Marco Zompit, M. et al. The CIP2A–TOPBP1 complex safeguards chromosomal stability during mitosis. Nat. Commun. 13, 4143 (2022).
Krupina, K., Goginashvili, A. & Cleveland, D. W. Scrambling the genome in cancer: causes and consequences of complex chromosome rearrangements. Nat. Rev. Genet. 25, 196–210 (2024).
Kim, H. et al. Extrachromosomal DNA is associated with oncogene amplification and poor outcome across multiple cancers. Nat. Genet. 52, 891–897 (2020).
Hall-Younger, E. & Tait, S. W. Mitochondria and cell death signalling. Curr. Opin. Cell Biol. 94, 102510 (2025).
Pizzuto, M. & Pelegrin, P. Cardiolipin in immune signaling and cell death. Trends Cell Biol. 30, 892–903 (2020).
Kalkavan, H. et al. Sublethal cytochrome c release generates drug-tolerant persister cells. Cell 185, 3356–3374 (2022).
Vringer, E. et al. Mitochondrial outer membrane integrity regulates a ubiquitin-dependent and NF-κB-mediated inflammatory response. EMBO J. 43, 904–930 (2024).
Di Bona, M. et al. Micronuclear collapse from oxidative damage. Science 385, eadj8691 (2024).
Martin, S. et al. A p62-dependent rheostat dictates micronuclei catastrophe and chromosome rearrangements. Science 385, eadj7446 (2024).
Garcia-Carpio, I. et al. Extra centrosomes induce PIDD1-mediated inflammation and immunosurveillance. EMBO J. 42, e113510 (2023).
Wang, R. W., Vigano, S., Ben-David, U., Amon, A. & Santaguida, S. Aneuploid senescent cells activate NF-κB to promote their immune clearance by NK cells. EMBO Rep. 22, e52032 (2021).
Xian, S. et al. The unfolded protein response links tumor aneuploidy to local immune dysregulation. EMBO Rep. 22, e52509 (2021).
Grootjans, J., Kaser, A., Kaufman, R. J. & Blumberg, R. S. The unfolded protein response in immunity and inflammation. Nat. Rev. Immunol. 16, 469–484 (2016).
Ahn, J. et al. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 5, 5166 (2014).
Chen, Q. et al. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533, 493–498 (2016).
Arwert, E. N. et al. STING and IRF3 in stromal fibroblasts enable sensing of genomic stress in cancer cells to undermine oncolytic viral therapy. Nat. Cell Biol. 22, 758–766 (2020).
Schadt, L. et al. Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity. Cell Rep. 29, 1236–1248 (2019).
Ahn, J., Xia, T., Rabasa Capote, A., Betancourt, D. & Barber, G. N. Extrinsic phagocyte-dependent STING signaling dictates the immunogenicity of dying cells. Cancer Cell 33, 862–873 (2018).
Dillon, M. T. et al. ATR inhibition potentiates the radiation-induced inflammatory tumor microenvironment. Clin. Cancer Res. 25, 3392–3403 (2019).
Sasaki, N. et al. RNA sensing induced by chromosome missegregation augments anti-tumor immunity. Mol. Cell 85, 770–786 (2025).
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).
Hustedt, N. & Durocher, D. The control of DNA repair by the cell cycle. Nat. Cell Biol. 19, 1–9 (2016).
Brambati, A. et al. RHINO directs MMEJ to repair DNA breaks in mitosis. Science 381, 653–660 (2023).
Rieder, C. L. & Cole, R. W. Entry into mitosis in vertebrate somatic cells is guarded by a chromosome damage checkpoint that reverses the cell cycle when triggered during early but not late prophase. J. Cell Biol. 142, 1013–1022 (1998).
Wang, H. et al. PLK1 targets CtIP to promote microhomology-mediated end joining. Nucleic Acids Res. 46, 10724–10739 (2018).
Martin, P. R. et al. The mitotic CIP2A–TOPBP1 axis facilitates mitotic pathway choice between MiDAS and MMEJ. Nat. Commun. 16, 10623 (2025).
Bhowmick, R., Minocherhomji, S. & Hickson, I. D. RAD52 facilitates mitotic DNA synthesis following replication stress. Mol. Cell 64, 1117–1126 (2016).
Mocanu, C. et al. DNA replication is highly resilient and persistent under the challenge of mild replication stress. Cell Rep. 39, 110701 (2022).
White, M. J. et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 (2014).
Frank, T. et al. Cell cycle arrest in mitosis promotes interferon-induced necroptosis. Cell Death Differ. 26, 2046–2060 (2019).
Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017).
Chen, K. W. & Broz, P. Gasdermins as evolutionarily conserved executors of inflammation and cell death. Nat. Cell Biol. 26, 1394–1406 (2024).
Gong, W. et al. Phosphorylated IRF3 promotes GSDME-mediated pyroptosis through RIPK1/FADD/caspase-8 complex formation during mitotic arrest in ovarian cancer. Cell Commun. Signal. 23, 306 (2025).
Hu, Y. et al. Paclitaxel induces micronucleation and activates pro-inflammatory cGAS–STING signaling in triple-negative breast cancer. Mol. Cancer Ther. 20, 2553–2567 (2021).
Scribano, C. M. et al. Chromosomal instability sensitizes patient breast tumors to multipolar divisions induced by paclitaxel. Sci. Transl. Med. 13, eabd4811 (2021).
Kayagaki, N. et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591, 131–136 (2021).
Acknowledgements
A.V. acknowledges support by the Austrian Science Fund (FWF) (10.55776/I6642, TRR353 and 10.55776/FG25) and the European Research Council (ERC AdG POLICE #787171). C.Z. acknowledges support from Cancer Research UK (RCCFEL\100092), the Cancer Research UK Radiation Research Centre of Excellence at The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust (A28724 and RRCOER-Jun24/100006) and Breast Cancer Now (2023.05PR1625). We also thank N. Kinz and M. Schapfl for help with preparing the figures.
Author information
Authors and Affiliations
Contributions
All authors contributed equally. D.R., C.Z. and A.V. led the reviewing and writing of individual sections that were subsequently cross-checked and edited by each author. C.Z. developed Fig. 1, A.V. developed Figs. 2–4. All figures were edited by D.R., C.Z. and A.V.
Corresponding authors
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.
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.
About this article
Cite this article
Rizzotto, D., Zierhut, C. & Villunger, A. Mitotic errors as triggers of cell death and inflammation. Nat Cell Biol 28, 21–34 (2026). https://doi.org/10.1038/s41556-025-01785-9
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41556-025-01785-9


