banner

News

Oct 17, 2024

Inflammasome protein scaffolds the DNA damage complex during tumor development | Nature Immunology

Nature Immunology (2024)Cite this article

163 Accesses

5 Altmetric

Metrics details

Inflammasome sensors activate cellular signaling machineries to drive inflammation and cell death processes. Inflammasomes also control the development of certain diseases independently of canonical functions. Here, we show that the inflammasome protein NLR family CARD domain-containing protein 4 (NLRC4) attenuated the development of tumors in the Apcmin/+ mouse model. This response was independent of inflammasome signaling by NLRP3, NLRP6, NLR family apoptosis inhibitory proteins, absent in melanoma 2, apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1 and caspase-11. NLRC4 interacted with the DNA-damage-sensing ataxia telangiectasia and Rad3-related (ATR)–ATR-interacting protein (ATRIP)–Ewing tumor-associated antigen 1 (ETAA1) complex to promote the recruitment of the checkpoint adapter protein claspin, licensing the activation of the kinase checkpoint kinase-1 (CHK1). Genotoxicity-induced activation of the NLRC4–ATR–ATRIP–ETAA1 complex drove the tumor-suppressing DNA damage response and CHK1 activation, and further attenuated the accumulation of DNA damage. These findings demonstrate a noninflammatory function of an inflammasome protein in promoting the DNA damage response and mediating protection against cancer.

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

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

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

All data are available in the main figures, Supplementary Information, extended data figures or source data. The raw microbiota (available from https://doi.org/10.5281/zenodo.10278738)124 and phosphoproteomics (available from https://doi.org/10.5281/zenodo.10278713)125 data have been deposited to Zenodo. Source data are provided with this paper.

Barnett, K. C., Li, S., Liang, K. & Ting, J. P. A 360 degrees view of the inflammasome: mechanisms of activation, cell death, and diseases. Cell 186, 2288–2312 (2023).

Article CAS PubMed PubMed Central Google Scholar

Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420 (2016).

Article CAS PubMed Google Scholar

Lamkanfi, M. & Dixit, V. M. In Retrospect: the inflammasome turns 15. Nature 548, 534–535 (2017).

Article CAS PubMed Google Scholar

Humphries, F. & Fitzgerald, K. A. Igniting the firestorm: the inflammasome in autoinflammatory syndromes. J. Allergy Clin. Immunol. 148, 1470–1472 (2021).

Article PubMed Google Scholar

Bauernfeind, F. & Hornung, V. Of inflammasomes and pathogens—sensing of microbes by the inflammasome. EMBO Mol. Med. 5, 814–826 (2013).

Article CAS PubMed PubMed Central Google Scholar

Heneka, M. T., McManus, R. M. & Latz, E. Inflammasome signalling in brain function and neurodegenerative disease. Nat. Rev. Neurosci. 19, 610–621 (2018).

Article CAS PubMed Google Scholar

Evavold, C. L. & Kagan, J. C. Inflammasomes: threat-assessment organelles of the innate immune system. Immunity 51, 609–624 (2019).

Article CAS PubMed PubMed Central Google Scholar

de Zoete, M. R., Palm, N. W., Zhu, S. & Flavell, R. A. Inflammasomes. Cold Spring Harb. Perspect. Biol. 6, a016287 (2014).

Article PubMed PubMed Central Google Scholar

Man, S. M., Karki, R. & Kanneganti, T. D. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol. Rev. 277, 61–75 (2017).

Article CAS PubMed PubMed Central Google Scholar

Zhao, Y. & Shao, F. Diverse mechanisms for inflammasome sensing of cytosolic bacteria and bacterial virulence. Curr. Opin. Microbiol. 29, 37–42 (2016).

Article PubMed Google Scholar

Xu, J. & Núñez, G. The NLRP3 inflammasome: activation and regulation. Trends Biochem. Sci. 48, 331–344 (2023).

Article CAS PubMed Google Scholar

Sharma, B. R. & Kanneganti, T.-D. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 22, 550–559 (2021).

Article CAS PubMed PubMed Central Google Scholar

Miao, E. A. et al. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1β via Ipaf. Nat. Immunol. 7, 569–575 (2006).

Article CAS PubMed Google Scholar

Miao, E. A., Ernst, R. K., Dors, M., Mao, D. P. & Aderem, A. Pseudomonas aeruginosa activates caspase 1 through Ipaf. Proc. Natl Acad. Sci. USA 105, 2562–2567 (2008).

Article CAS PubMed PubMed Central Google Scholar

Franchi, L. et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1β in salmonella-infected macrophages. Nat. Immunol. 7, 576–582 (2006).

Article CAS PubMed Google Scholar

Kofoed, E. M. & Vance, R. E. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity. Nature 477, 592–595 (2011).

Article CAS PubMed PubMed Central Google Scholar

Zhao, Y. et al. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature 477, 596–600 (2011).

Article CAS PubMed Google Scholar

Bürckstümmer, T. et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat. Immunol. 10, 266–272 (2009).

Article PubMed Google Scholar

Hornung, V. et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458, 514–518 (2009).

Article CAS PubMed PubMed Central Google Scholar

Roberts, T. L. et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science 323, 1057–1060 (2009).

Article CAS PubMed Google Scholar

Fernandes-Alnemri, T., Yu, J. W., Datta, P., Wu, J. & Alnemri, E. S. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458, 509–513 (2009).

Article CAS PubMed PubMed Central Google Scholar

Cerretti, D. P. et al. Molecular cloning of the interleukin-1β converting enzyme. Science 256, 97–100 (1992).

Article CAS PubMed Google Scholar

Thornberry, N. A. et al. A novel heterodimeric cysteine protease is required for interleukin-1β processing in monocytes. Nature 356, 768–774 (1992).

Article CAS PubMed Google Scholar

Ghayur, T. et al. Caspase-1 processes IFN-γ-inducing factor and regulates LPS-induced IFN-γ production. Nature 386, 619–623 (1997).

Article CAS PubMed Google Scholar

Gu, Y. et al. Activation of interferon-γ inducing factor mediated by interleukin-1β converting enzyme. Science 275, 206–209 (1997).

Article CAS PubMed Google Scholar

Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015).

Article CAS PubMed Google Scholar

Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

Article CAS PubMed Google Scholar

Shi, X. et al. Recognition and maturation of IL-18 by caspase-4 noncanonical inflammasome. Nature 624, 442–450 (2023).

Article CAS PubMed Google Scholar

Devant, P. et al. Structural insights into cytokine cleavage by inflammatory caspase-4. Nature 624, 451–459 (2023).

Article CAS PubMed PubMed Central Google Scholar

Heilig, R. et al. The Gasdermin-D pore acts as a conduit for IL-1β secretion in mice. Eur. J. Immunol. 48, 584–592 (2018).

Article CAS PubMed Google Scholar

Evavold, C. L. et al. The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages. Immunity 48, 35–44 (2018).

Article CAS PubMed Google Scholar

Xia, S. et al. Gasdermin D pore structure reveals preferential release of mature interleukin-1. Nature 593, 607–611 (2021).

Article CAS PubMed PubMed Central Google Scholar

Chan, A. H. & Schroder, K. Inflammasome signaling and regulation of interleukin-1 family cytokines. J. Exp. Med. 217, e20190314 (2020).

Article PubMed Google Scholar

Zengeler, K. E. & Lukens, J. R. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat. Rev. Immunol. 21, 454–468 (2021).

Article CAS PubMed Google Scholar

Pandey, A., Shen, C., Feng, S. & Man, S. M. Cell biology of inflammasome activation. Trends Cell Biol. 31, 924–939 (2021).

Article CAS PubMed Google Scholar

Man, S. M. et al. Salmonella infection induces recruitment of Caspase-8 to the inflammasome to modulate IL-1β production. J. Immunol. 191, 5239–5246 (2013).

Article CAS PubMed Google Scholar

Gurung, P. et al. FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J. Immunol. 192, 1835–1846 (2014).

Article CAS PubMed Google Scholar

Vince, J. E. et al. The mitochondrial apoptotic effectors BAX/BAK activate caspase-3 and -7 to trigger NLRP3 inflammasome and caspase-8 driven IL-1β activation. Cell Rep. 25, 2339–2353 (2018).

Article CAS PubMed Google Scholar

Doerflinger, M. et al. Flexible usage and interconnectivity of diverse cell death pathways protect against intracellular infection. Immunity 53, 533–547 (2020).

Article CAS PubMed Google Scholar

Roncaioli, J. L. et al. A hierarchy of cell death pathways confers layered resistance to shigellosis in mice. eLife 12, e83639 (2023).

Article CAS PubMed PubMed Central Google Scholar

Christgen, S. et al. Identification of the PANoptosome: a molecular platform triggering pyroptosis, apoptosis, and necroptosis (PANoptosis). Front. Cell. Infect. Microbiol. 10, 237 (2020).

Article CAS PubMed PubMed Central Google Scholar

Lee, S. et al. AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence. Nature 597, 415–419 (2021).

Article CAS PubMed PubMed Central Google Scholar

Sundaram, B. et al. NLRP12-PANoptosome activates PANoptosis and pathology in response to heme and PAMPs. Cell 186, 2783–2801 (2023).

Article CAS PubMed PubMed Central Google Scholar

Moser, A. R., Pitot, H. C. & Dove, W. F. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 247, 322–324 (1990).

Article CAS PubMed Google Scholar

Su, L. K. et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 256, 668–670 (1992).

Article CAS PubMed Google Scholar

Canna, S. W. et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat. Genet. 46, 1140–1146 (2014).

Article CAS PubMed PubMed Central Google Scholar

Romberg, N. et al. Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation. Nat. Genet. 46, 1135–1139 (2014).

Article CAS PubMed Google Scholar

Kitamura, A., Sasaki, Y., Abe, T., Kano, H. & Yasutomo, K. An inherited mutation in NLRC4 causes autoinflammation in human and mice. J. Exp. Med. 211, 2385–2396 (2014).

Article CAS PubMed Google Scholar

Moghaddas, F. et al. Autoinflammatory mutation in NLRC4 reveals a leucine-rich repeat (LRR)-LRR oligomerization interface. J. Allergy Clin. Immunol. 142, 1956–1967 (2018).

Article CAS PubMed Google Scholar

Steiner, A. et al. Recessive NLRC4-autoinflammatory disease reveals an ulcerative colitis locus. J. Clin. Immunol. 42, 325–335 (2022).

Article CAS PubMed Google Scholar

Nichols, R. D., von Moltke, J. & Vance, R. E. NAIP/NLRC4 inflammasome activation in MRP8+ cells is sufficient to cause systemic inflammatory disease. Nat. Commun. 8, 2209 (2017).

Article PubMed PubMed Central Google Scholar

Lee, S. H. et al. ERK activation drives intestinal tumorigenesis in Apcmin/+ mice. Nat. Med. 16, 665–670 (2010).

Article CAS PubMed PubMed Central Google Scholar

Zhang, L. et al. Cryo-EM structure of the activated NAIP2-NLRC4 inflammasome reveals nucleated polymerization. Science 350, 404–409 (2015).

Article CAS PubMed PubMed Central Google Scholar

Hu, Z. et al. Structural and biochemical basis for induced self-propagation of NLRC4. Science 350, 399–404 (2015).

Article CAS PubMed Google Scholar

Paidimuddala, B. et al. Mechanism of NAIP-NLRC4 inflammasome activation revealed by cryo-EM structure of unliganded NAIP5. Nat. Struct. Mol. Biol. 30, 159–166 (2023).

Article CAS PubMed PubMed Central Google Scholar

Paidimuddala, B., Cao, J. & Zhang, L. Structural basis for flagellin-induced NAIP5 activation. Sci. Adv. 9, eadi8539 (2023).

Article CAS PubMed PubMed Central Google Scholar

Ayres, J. S., Trinidad, N. J. & Vance, R. E. Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota. Nat. Med. 18, 799–806 (2012).

Article CAS PubMed PubMed Central Google Scholar

Rauch, I. et al. NAIP proteins are required for cytosolic detection of specific bacterial ligands in vivo. J. Exp. Med. 213, 657–665 (2016).

Article CAS PubMed PubMed Central Google Scholar

Zhao, Y. et al. Genetic functions of the NAIP family of inflammasome receptors for bacterial ligands in mice. J. Exp. Med. 213, 647–656 (2016).

Article CAS PubMed PubMed Central Google Scholar

Mitchell, P. S. et al. NAIP–NLRC4-deficient mice are susceptible to shigellosis. eLife 9, e59022 (2020).

Article CAS PubMed PubMed Central Google Scholar

Allam, R. et al. Epithelial NAIPs protect against colonic tumorigenesis. J. Exp. Med. 212, 369–383 (2015).

Article CAS PubMed PubMed Central Google Scholar

Hu, B. et al. Inflammation-induced tumorigenesis in the colon is regulated by caspase-1 and NLRC4. Proc. Natl Acad. Sci. USA 107, 21635–21640 (2010).

Article PubMed PubMed Central Google Scholar

Kolb, R. et al. Obesity-associated NLRC4 inflammasome activation drives breast cancer progression. Nat. Commun. 7, 13007 (2016).

Article CAS PubMed PubMed Central Google Scholar

Ohashi, K. et al. NOD-like receptor C4 inflammasome regulates the growth of colon cancer liver metastasis in NAFLD. Hepatology 70, 1582–1599 (2019).

Article CAS PubMed Google Scholar

Kumar, Y., Radha, V. & Swarup, G. Interaction with Sug1 enables Ipaf ubiquitination leading to caspase 8 activation and cell death. Biochem. J. 427, 91–104 (2010).

Article CAS PubMed Google Scholar

Man, S. M. et al. Inflammasome activation causes dual recruitment of NLRC4 and NLRP3 to the same macromolecular complex. Proc. Natl Acad. Sci. USA 111, 7403–7408 (2014).

Article CAS PubMed PubMed Central Google Scholar

Rauch, I. et al. NAIP-NLRC4 inflammasomes coordinate intestinal epithelial cell expulsion with eicosanoid and IL-18 release via activation of caspase-1 and -8. Immunity 46, 649–659 (2017).

Article CAS PubMed PubMed Central Google Scholar

Lee, B. L. et al. ASC- and caspase-8-dependent apoptotic pathway diverges from the NLRC4 inflammasome in macrophages. Sci. Rep. 8, 3788 (2018).

Article PubMed PubMed Central Google Scholar

Kay, C., Wang, R., Kirkby, M. & Man, S. M. Molecular mechanisms activating the NAIP‐NLRC4 inflammasome: implications in infectious disease, autoinflammation, and cancer. Immunol. Rev. 297, 67–82 (2020).

Article CAS PubMed Google Scholar

Man, S. M. & Jenkins, B. J. Context-dependent functions of pattern recognition receptors in cancer. Nat. Rev. Cancer 22, 397–413 (2022).

Article CAS PubMed Google Scholar

MacDonald, B. T., Tamai, K. & He, X. Wnt/β-catenin signaling: components, mechanisms, and diseases. Dev. Cell 17, 9–26 (2009).

Article CAS PubMed PubMed Central Google Scholar

Stamos, J. L. & Weis, W. I. The β-catenin destruction complex. Cold Spring Harb. Perspect. Biol. 5, a007898 (2013).

Article PubMed PubMed Central Google Scholar

Shih, I. M. et al. Top-down morphogenesis of colorectal tumors. Proc. Natl Acad. Sci. USA 98, 2640–2645 (2001).

Article CAS PubMed PubMed Central Google Scholar

Mah, L., El-Osta, A. & Karagiannis, T. γH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 24, 679–686 (2010).

Article CAS PubMed Google Scholar

Pandey, A. et al. Ku70 senses cytosolic DNA and assembles a tumor-suppressive signalosome. Sci. Adv. 10, eadh3409 (2024).

Article CAS PubMed PubMed Central Google Scholar

Montecucco, A. & Biamonti, G. Cellular response to etoposide treatment. Cancer Lett. 252, 9–18 (2007).

Article CAS PubMed Google Scholar

Giglia-Mari, G., Zotter, A. & Vermeulen, W. DNA damage response. Cold Spring Harb. Perspect. Biol. 3, a000745 (2011).

Article PubMed PubMed Central Google Scholar

Zhao, H. & Piwnica-Worms, H. ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1. Mol. Cell. Biol. 21, 4129–4139 (2001).

Article CAS PubMed PubMed Central Google Scholar

Jiang, K. et al. Regulation of Chk1 includes chromatin association and 14-3-3 binding following phosphorylation on Ser-345. J. Biol. Chem. 278, 25207–25217 (2003).

Article CAS PubMed Google Scholar

Karlsson-Rosenthal, C. & Millar, J. B. A. Cdc25: mechanisms of checkpoint inhibition and recovery. Trends Cell Biol. 16, 285–292 (2006).

Article CAS PubMed Google Scholar

Busino, L., Chiesa, M., Draetta, G. F. & Donzelli, M. Cdc25A phosphatase: combinatorial phosphorylation, ubiquitylation and proteolysis. Oncogene 23, 2050–2056 (2004).

Article CAS PubMed Google Scholar

Blackford, A. N. & Jackson, S. P. ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response. Mol. Cell 66, 801–817 (2017).

Article CAS PubMed Google Scholar

Wang, C. et al. Inducing and exploiting vulnerabilities for the treatment of liver cancer. Nature 574, 268–272 (2019).

Article CAS PubMed PubMed Central Google Scholar

Poyet, J. L. et al. Identification of Ipaf, a human caspase-1-activating protein related to Apaf-1. J. Biol. Chem. 276, 28309–28313 (2001).

Article CAS PubMed Google Scholar

Damiano, J. S., Stehlik, C., Pio, F., Godzik, A. & Reed, J. C. CLAN, a novel human CED-4-like gene. Genomics 75, 77–83 (2001).

Article CAS PubMed Google Scholar

Lecona, E. & Fernandez-Capetillo, O. Targeting ATR in cancer. Nat. Rev. Cancer 18, 586–595 (2018).

Article CAS PubMed Google Scholar

Lin, S.-Y., Liang, Y. & Li, K. Multiple roles of BRIT1/MCPH1 in DNA damage response, DNA repair, and cancer suppression. Yonsei Med. J. 51, 295–301 (2010).

Article CAS PubMed PubMed Central Google Scholar

Kumagai, A. & Dunphy, W. G. Claspin, a novel protein required for the activation of Chk1 during a DNA replication checkpoint response in Xenopus egg extracts. Mol. Cell 6, 839–849 (2000).

Article CAS PubMed Google Scholar

Aghabozorgi, A. S. et al. Role of adenomatous polyposis coli (APC) gene mutations in the pathogenesis of colorectal cancer; current status and perspectives. Biochimie 157, 64–71 (2019).

Article CAS PubMed Google Scholar

Becker, W. R. et al. Single-cell analyses define a continuum of cell state and composition changes in the malignant transformation of polyps to colorectal cancer. Nat. Genet. 54, 985–995 (2022).

Article CAS PubMed PubMed Central Google Scholar

Lonsdale, J. et al. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

Article CAS Google Scholar

Tang, Z., Kang, B., Li, C., Chen, T. & Zhang, Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47, W556–W560 (2019).

Article CAS PubMed PubMed Central Google Scholar

Janowski, A. M. et al. NLRC4 suppresses melanoma tumor progression independently of inflammasome activation. J. Clin. Invest. 126, 3917–3928 (2016).

Article PubMed PubMed Central Google Scholar

Tubbs, A. & Nussenzweig, A. Endogenous DNA damage as a source of genomic instability in cancer. Cell 168, 644–656 (2017).

Article CAS PubMed PubMed Central Google Scholar

Negrini, S., Gorgoulis, V. G. & Halazonetis, T. D. Genomic instability—an evolving hallmark of cancer. Mol. Cell Biol. 11, 220–228 (2010).

CAS Google Scholar

Bartek, J., Bartkova, J. & Lukas, J. DNA damage signalling guards against activated oncogenes and tumour progression. Oncogene 26, 7773–7779 (2007).

Article CAS PubMed Google Scholar

Fang, Y. et al. ATR functions as a gene dosage‐dependent tumor suppressor on a mismatch repair‐deficient background. EMBO J. 23, 3164–3174 (2004).

Article CAS PubMed PubMed Central Google Scholar

Liu, Q. et al. Chk1 is an essential kinase that is regulated by Atr and required for the G2/M DNA damage checkpoint. Genes Dev. 14, 1448–1459 (2000).

Article CAS PubMed PubMed Central Google Scholar

Greenow, K. R., Clarke, A. R., Williams, G. T. & Jones, R. Wnt-driven intestinal tumourigenesis is suppressed by Chk1 deficiency but enhanced by conditional haploinsufficiency. Oncogene 33, 4089–4096 (2014).

Article CAS PubMed Google Scholar

Cortez, D., Guntuku, S., Qin, J. & Elledge, S. J. ATR and ATRIP: partners in checkpoint signaling. Science 294, 1713–1716 (2001).

Article CAS PubMed Google Scholar

Zhang, R. et al. NLRC4 promotes the cGAS‐STING signaling pathway by facilitating CBL‐mediated K63‐linked polyubiquitination of TBK1. J. Med. Virol. 95, e29013 (2023).

Article CAS PubMed Google Scholar

Hu, B. et al. The DNA-sensing AIM2 inflammasome controls radiation-induced cell death and tissue injury. Science 354, 765–768 (2016).

Article CAS PubMed PubMed Central Google Scholar

Lammert, C. R. et al. AIM2 inflammasome surveillance of DNA damage shapes neurodevelopment. Nature 580, 647–652 (2020).

Article CAS PubMed PubMed Central Google Scholar

Bodnar-Wachtel, M. et al. Inflammasome-independent NLRP3 function enforces ATM activity in response to genotoxic stress. Life Sci. Alliance 6, e202201494 (2023).

Article CAS PubMed PubMed Central Google Scholar

Jones, J. W. et al. Absent in melanoma 2 is required for innate immune recognition of Francisella tularensis. Proc. Natl Acad. Sci. USA 107, 9771–9776 (2010).

Article CAS PubMed PubMed Central Google Scholar

Mariathasan, S. et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430, 213–218 (2004).

Article CAS PubMed Google Scholar

Kuida, K. et al. Altered cytokine export and apoptosis in mice deficient in interleukin-1β converting enzyme. Science 267, 2000–2003 (1995).

Article CAS PubMed Google Scholar

Wang, S. et al. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell 92, 501–509 (1998).

Article CAS PubMed Google Scholar

Kovarova, M. et al. NLRP1-dependent pyroptosis leads to acute lung injury and morbidity in mice. J. Immunol. 189, 2006–2016 (2012).

Article CAS PubMed Google Scholar

Zheng, D. et al. Epithelial Nlrp10 inflammasome mediates protection against intestinal autoinflammation. Nat. Immunol. 24, 585–594 (2023).

Article CAS PubMed Google Scholar

Hu, Q. et al. Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat. Immunol. 20, 835–851 (2019).

Article CAS PubMed PubMed Central Google Scholar

Medhavy, A. et al. A TNIP1-driven systemic autoimmune disorder with elevated IgG4. Nat. Immunol. 25, 1678–1691 (2024).

Article CAS PubMed PubMed Central Google Scholar

Karki, R. et al. NLRC3 is an inhibitory sensor of PI3K-mTOR pathways in cancer. Nature 540, 583–587 (2016).

Article CAS PubMed PubMed Central Google Scholar

Man, S. M. et al. Critical role for the DNA sensor AIM2 in stem cell proliferation and cancer. Cell 162, 45–58 (2015).

Article CAS PubMed PubMed Central Google Scholar

Jing, W. et al. Clostridium septicum α-toxin activates the NLRP3 inflammasome by engaging GPI-anchored proteins. Sci. Immunol. 7, eabm1803 (2022).

Article CAS PubMed Google Scholar

Fox, D. et al. Bacillus cereus non-haemolytic enterotoxin activates the NLRP3 inflammasome. Nat. Commun. 11, 760 (2020).

Article CAS PubMed PubMed Central Google Scholar

Luu, L. D. W., Zhong, L., Kaur, S., Raftery, M. J. & Lan, R. Comparative phosphoproteomics of classical Bordetellae elucidates the potential role of serine, threonine and tyrosine phosphorylation in Bordetella biology and virulence. Front. Cell. Infect. Microbiol. 11, 660280 (2021).

Article CAS PubMed PubMed Central Google Scholar

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

Mathur, A. et al. A multicomponent toxin from Bacillus cereus incites inflammation and shapes host outcome via the NLRP3 inflammasome. Nat. Microbiol. 4, 362–374 (2019).

Article CAS PubMed Google Scholar

Feng, S. et al. Pathogen-selective killing by guanylate-binding proteins as a molecular mechanism leading to inflammasome signaling. Nat. Commun. 13, 4395 (2022).

Article CAS PubMed PubMed Central Google Scholar

Caporaso, J. G. et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl Acad. Sci. USA 108, 4516–4522 (2011).

Article CAS PubMed Google Scholar

Deshpande, N. P., Riordan, S. M., Castaño-Rodríguez, N., Wilkins, M. R. & Kaakoush, N. O. Signatures within the esophageal microbiome are associated with host genetics, age, and disease. Microbiome 6, 227 (2018).

Article PubMed PubMed Central Google Scholar

Yang, L. & Chen, J. A comprehensive evaluation of microbial differential abundance analysis methods: current status and potential solutions. Microbiome 10, 130 (2022).

Article CAS PubMed PubMed Central Google Scholar

Kaakoush, N. O. & Man, S. M. Fecal microbiota profiling of NLRC4 KO mice and their WT littermates. Zenodo https://zenodo.org/records/10278738 (2023).

Kaakoush, N. O. & Man, S. M. Phosphoproteomics of NLRC4 KO mice and wild-type littermates. Zenodo https://doi.org/10.5281/zenodo.10278713 (2023).

Download references

We thank the facilities and scientific and technical assistance of Microscopy Australia at the Centre for Advanced Microscopy (ANU), which is funded by the University and the Federal Government. We thank the National Collaborative Research Infrastructure Strategy (NCRIS) via Phenomics Australia. We thank the South Australian Genomics Centre (SAGC), which is supported by NCRIS via BioPlatforms Australia and by the SAGC partner institutes. We thank the facilities and scientific and technical assistance of the Cytometry, Histology and Spatial Multiomics facility of the John Curtin School of Medical Research, ANU. We thank G. Burgio (ANU) for generating the Nlrp6−/− mice, R. E. Vance and E. Turcotte (University of California) for providing the WT and NLRC4−/− THP-1 cell lines, and J.-W. Yu (Yonsei University College of Medicine) for providing the anti-NLRC4 antibody. We thank members of the Man laboratory for their comments and suggestions; the National Health and Medical Research Council of Australia under project grant no. APP1146864, an ideas grant no. APP2002686 and an investigator grant no. 2026910 (S.M.M.); a CSL Centenary Fellowship (S.M.M.); a John Curtin School of Medical Research PhD Scholarship (C.S.); the Gretel and Gordon Bootes Medical Research Foundation (C.S.); a Cancer Council ACT Research Grant (A.M.); a Gastroenterological Society of Australia GESA Mostyn Family Grant (D.E.T.); and an Australian Research Council Centre of Excellence for the Mathematical Analysis of Cellular Systems grant no. CE230100001 (L.L., H.Y., J.W.).

Division of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia

Cheng Shen, Abhimanu Pandey, Daniel Enosi Tuipulotu, Anukriti Mathur, Nilanthi K. Adikari, Chinh Ngo, Weidong Jing, Shouya Feng, Yuwei Hao, Anyang Zhao, Max Kirkby, Melan Kurera, Jing Zhang, Shweta Venkataraman & Si Ming Man

Division of Genome Sciences and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia

Lixinyu Liu, Haoyu Yang & Jiayu Wen

ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Canberra, Australian Capital Territory, Australia

Lixinyu Liu, Haoyu Yang & Jiayu Wen

Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia

Cheng Liu

School of Medicine, University of Queensland, Herston, Queensland, Australia

Cheng Liu

Mater Pathology, Mater Hospital, South Brisbane, Queensland, Australia

Cheng Liu

Epigenetics and RNA Biology Laboratory, The School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia

Renhua Song & Justin J.-L. Wong

The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia

Ulrike Schumann & Riccardo Natoli

The Shine Dalgarno Centre for RNA Innovation, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia

Ulrike Schumann & Riccardo Natoli

The Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia

Ulrike Schumann

School of Medicine and Psychology, The Australian National University, Canberra, Australian Capital Territory, Australia

Riccardo Natoli

Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA

Liman Zhang

School of Biomedical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia

Nadeem O. Kaakoush

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

C.S. and S.M.M. conceptualized the study. C.S., A.P., D.E.T., L.L., H.Y., C.N., Y.H., R.S. and N.O.K. devised the methodology. C.S., A.P., D.E.T., A.M., L.L., H.Y., N.K.A., C.N., W.J., S.F., Y.H., A.Z., Max Kirkby, Melan Kurera, J.Z., S.V., C.L., R.S., U.S. and N.O.K. carried out the investigation. C.S., A.P., D.E.T., A.M., L.L., H.Y., C.L., R.S., J.J.-L.W., R.N., J.W., L.Z., N.O.K. and S.M.M. carried out the formal analysis. S.M.M. acquired the funding. C.S. and S.M.M. managed the project. S.M.M. supervised the study. C.S. and S.M.M. wrote the original manuscript draft. All authors reviewed and edited the manuscript draft.

Correspondence to Si Ming Man.

The authors declare no competing interests.

Nature Immunology thanks Tyler Curiel, Khashayarsha Khazaie and Igor Brodsky for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey in collaboration with the Nature Immunology editorial team. Peer reviewer reports are available.

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

a, Body weight of 20-week-old Apcmin/+ (n = 12) and Apcmin/+ Nlrc4–/– mice (n = 12), and small intestine and colon length of 20-week-old Apcmin/+ (n = 13) and Apcmin/+ Nlrc4–/– mice (n = 14). b, Species richness, species evenness and Shannon’s diversity of the microbiome in the small intestine of 20-week-old Apcmin/+ (n = 10) and Apcmin/+ Nlrc4–/– mice (n = 14). c, Principal coordinate analysis and differential abundance analysis for the microbiome in the small intestine of 20-week-old Apcmin/+ (n = 10) and Apcmin/+ Nlrc4–/– mice (n = 14). ANOSIM (analysis of similarity, R = −0.011, P = 0.52). q indicates an adjusted p-value. d, Number and size of tumors in the colon of 20-week-old Apcmin/+ (n = 13) and Apcmin/+ Nlrc4–/– mice (n = 14). Each symbol represents one individual mouse (a–d). Data represent mean ± SEM; two-tailed t-test (a, b and d).

Source data

a–d, Number and size of tumors in the small intestine and colon of 20-week-old Apcmin/+ (n = 13) and Apcmin/+ Aim2–/– mice (n = 15) (a); Apcmin/+ (n = 14) and Apcmin/+ Casp11–/– mice (n = 17) (b); Apcmin/+ (n = 11) and Apcmin/+ Nlrp3–/– mice (n = 13) (c); Apcmin/+ (n = 14) and Apcmin/+ Nlrp6–/– mice (n = 12) (d). Each symbol represents one individual mouse (a–d). Data represent mean ± SEM; two-tailed t-test (a–d).

Source data

a, Densitometric quantification of Casp-1 p20 and GSDMD p30 in the small intestine of Apcmin/+ (n = 6) and Apcmin/+ Nlrc4–/– (n = 6) mice in Fig. 2c. b, Number and size of tumors in the small intestine and colon of 10-week-old Apcmin/+ (n = 15) and Apcmin/+ Nlrc4–/– mice (n = 13). c,d, Number and size of tumors in the small intestine of 20-week-old Apcmin/+ (n = 14) and Apcmin/+ Asc–/– mice (n = 12) (c); Apcmin/+ (n = 12) and Apcmin/+ Casp1–/– mice (n = 12) (d). e,f, Number and size of tumors in the colon of 20-week-old Apcmin/+ (n = 14) and Apcmin/+ Asc–/– mice (n = 12) (e); Apcmin/+ (n = 12) and Apcmin/+ Casp1–/– mice (n = 12) (f). Each symbol represents one individual mouse (a–f). Data represent mean ± SEM; two-tailed t-test (a–f).

Source data

a, Densitometric quantification of β-catenin in Fig. 4a (cytosolic β-catenin was normalized to GAPDH; nuclear β-catenin was normalized to Lamin B1). b,c, Quantification (b) and representative images (c) of nuclear β-catenin in the small intestine of 10-week-old Apcmin/+ (n = 5) and Apcmin/+ Nlrc4–/– mice (n = 5) (each symbol represents one microscopy image). Scale bar for all images, 50 μm. d, Relative gene expression of Axin2, Lgr5 and Tcf7 in healthy tissues from the small intestine of 10-week-old Apc+/+ mice, and in tumor tissues from the small intestine of 10-week-old Apcmin/+ mice. (normalized to Gapdh). e, Relative gene expression of Axin2, Lgr5 and Tcf7 in the small intestine of 10-week-old Apcmin/+ and Apcmin/+ Nlrc4–/– mice (normalized to Gapdh). f, Levels of differential phosphorylated proteins (Supplementary Table 1, P < 0.05) in the small intestine of 20-week-old Apcmin/+ (n = 5) and Apcmin/+ Nlrc4–/– mice (n = 5). g,h, Representative images (g) and quantification (h) of γH2AX in Apc+/+ and Apc+/+ Nlrc4–/– BMDMs treated with a vehicle control or 50 µM etoposide for 20 h (each symbol represents one microscopy image). Scale bar, 40 μm. Image capture and quantification were performed in a blinded manner (b and h). Each symbol or lane represents one individual mouse (a, d, e and f). Data are pooled from multiple Apcmin/+ (n = 5) and Apcmin/+ Nlrc4–/– (n = 5) mice (b); Apc+/+ (n = 5) and Apcmin/+ (n = 4) mice (d); Apcmin/+ (n = 8) and Apcmin/+ Nlrc4–/– (n = 8) mice (e), or from two independent experiments (h). Data represent mean ± SEM; two-tailed t-test (a, b, d, e and h).

Source data

a,b, Immunoblot analysis (a) and densitometric quantification (b) of p-BRCA1 S1524, p-p53 S15 and p-CHK2 T68 in the small intestine of 10-week-old Apcmin/+ (n = 6) and Apcmin/+ Nlrc4–/– (n = 6) mice (normalized to β-actin in Fig. 5a). c, Immunoblot analysis of NLRC4, p-CHK1 S345, CHK1, and β-actin in Apc+/+ and Apc+/+ Nlrc4–/– BMDMs treated with 50 µM etoposide for the indicated time. d, Immunoblot analysis of NLRC4, p-CHK1 S345, CHK1, and β-actin in Apc+/+ and Apc+/+ Nlrc4–/– BMDMs either left untreated or treated with 6 Gy ionizing radiation, followed by incubation for the indicated time. e, Immunoblot analysis of p-CHK1 S345, CHK1, and β-actin in WT and NLRC4–/– THP-1 cells treated with 10 mM hydrogen peroxide (H2O2) for the indicated time. f–h, Densitometric quantification of p-CHK1 S345 against CHK1 in the blots (f is related to d); (g is related to Fig. 5d); (h is related to e). i,j, Immunoblot analysis (i) and densitometric quantification (j) of p-CHK1 S345, CHK1, p-CDC25A S124, CDC25A, and β-actin in the small intestine of 10-week-old Apc+/+ (n = 6) and Apc+/+ Nlrc4–/– (n = 6) mice. Data represent one of three independent experiments (c–h). Each symbol or lane represents one individual mouse (a, b, i and j). Data represent mean ± SEM; two-tailed t-test (b and j).

Source data

a, Immunoblot analysis of GFP-NLRC4, ATR, and β-actin following immunoprecipitation of control IgG or anti-ATR antibody in cell lysates of GFP-NLRC4-expressing HEK239T cells. b, Immunoblot analysis of NLRC4, p-CHK1 S345, CHK1 and β-actin in Apcmin/+ and Apcmin/+ Nlrc4–/– small intestinal organoids pre-treated with a vehicle control or 1 µM AZD6738 for 1 h, followed by 50 µM etoposide treatment for the indicated time. AZD6738-treated organoids were maintained under the condition of AZD6738 inhibition during etoposide treatment. c, Analysis of ROS levels in live Apc+/+ and Apc+/+ Nlrc4–/– BMDMs treated with phosphate-buffered saline (PBS), 50 µM etoposide, or 10 mM hydrogen peroxide (H2O2) for the indicated time. d,e, Immunoblot analysis (d) and densitometric quantification (e) of NLRC4, p-CHK1 S345, CHK1, and β-actin in BMDMs pre-treated with a vehicle control or 500 µM N-acetyl cysteine (NAC) for 20 h, followed by 50 µM etoposide treatment for the indicated time. f,g, Immunoblot analysis (f) and densitometric quantification (g) of p-ERK1/2 T202/Y204 and ERK1/2 in BMDMs pre-treated with a vehicle control or 500 µM N-acetyl cysteine (NAC) for 20 h, followed by 1 µg/ml lipopolysaccharide (LPS) treatment for the indicated time. NAC-treated cells were maintained under the condition of NAC inhibition during etoposide treatment (d–g). Data represent one of three independent experiments (a–g). Data represent mean ± SEM; Two-way ANOVA with Holm-Sidak’s multiple comparisons test (c).

Source data

a, Schematic representation of the full-length NLRC4 protein and truncated mutants. b, Densitometric quantification of p-CHK1 S345 against CHK1 in the blots of Fig. 6e. c, Schematic representation of the full-length ATR protein and truncated mutants. d, Immunoblot analysis of GFP-NLRC4, β-actin, and the full-length V5-ATR protein or V5-ATR lacking the HEAT domain (ΔHEAT), FAT domain (ΔFAT), PIKK domain (ΔPIKK), PRD domain (ΔPRD) or FATC domain (ΔFATC) following immunoprecipitation using a control IgG or anti-GFP antibody from the cell lysates of V5-ATR (WT, ΔHEAT, ΔFAT, ΔPIKK, ΔPRD or ΔFATC)- and GFP-NLRC4-expressing HEK239T cells. e, Densitometric quantification of Claspin in the small intestine lysates of 10-week-old Apcmin/+ (n = 6) and Apcmin/+ Nlrc4–/– (n = 6) mice, related to Fig. 6h, i. Data represent one of three independent experiments (d). Data represent mean ± SEM; two-tailed t-test (e).

Source data

a, Dot plot representation of human NLRC4 expression across different cell types during malignant transformation of colorectal cancer (CRC). Data were obtained from GSE201349.

a, Pearson correlation analysis of the gene expression between NLRC4 and ATR, ATRIP, CLSPN, CHEK1, and CDC25A in the human small intestine. b, KEGG pathway enrichment analysis of genes showing the highest Pearson correlation coefficients with NLRC4 in the human small intestine. Data were obtained from GTEx (Small Intestine – Terminal Ileum) (a, and b). Two-tailed Pearson correlation coefficient test (a).

Left, in response to genomic stress, such as DNA damage assault, NLRC4 interacts with the ATR-ATRIP-ETAA1 complex to promote the recruitment of Claspin, leading to CHK1 activation. Activated CHK1 phosphorylates CDC25A, resulting in the degradation of CDC25A, and triggers the DNA damage response. Sufficient DNA damage response provides a protective response against tumor development. Right, the lack of NLRC4 hinders the recruitment of Claspin to the ATR-ATRIP-ETAA1 complex, leading to impaired DNA damage response. This defect causes DNA damage accumulation, leading to genomic instability and increased tumor development. Figures are drawn using PowerPoint.

Supplementary Tables 1–4.

Supplementary Table 1: Phospho-MS data_(NLRC4 WT VS KO);Supplementary Table 2: Top NLRC4-related genes in the human small intestine; Supplementary Table 3: NLRP6 knockout mouse strain information; Supplementary Table 4: Antibodies and primers.

Statistical source data.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Statistical source data.

Statistical source data.

Statistical source data.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

Statistical source data.

Unprocessed immunoblots.

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

Shen, C., Pandey, A., Enosi Tuipulotu, D. et al. Inflammasome protein scaffolds the DNA damage complex during tumor development. Nat Immunol (2024). https://doi.org/10.1038/s41590-024-01988-6

Download citation

Received: 26 February 2024

Accepted: 13 September 2024

Published: 14 October 2024

DOI: https://doi.org/10.1038/s41590-024-01988-6

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

SHARE