Pyroptosis, metabolism, and tumor immune microenvironment
Tiantian Du
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorJie Gao
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorPeilong Li
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorYunshan Wang
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorQiuchen Qi
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorXiaoyan Liu
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorCorresponding Author
Juan Li
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Chuanxin Wang
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Lutao Du
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorTiantian Du
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorJie Gao
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorPeilong Li
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
These authors contributed equally to this work.
Search for more papers by this authorYunshan Wang
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorQiuchen Qi
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorXiaoyan Liu
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Search for more papers by this authorCorresponding Author
Juan Li
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Chuanxin Wang
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Lutao Du
Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
Shandong Engineering and Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
Correspondence
Lutao Du, Chuanxin Wang, and Juan Li, Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan 250033, Shandong, China.
Email: [email protected]; [email protected]; [email protected]
Search for more papers by this authorAbstract
In response to a wide range of stimulations, host cells activate pyroptosis, a kind of inflammatory cell death which is provoked by the cytosolic sensing of danger signals and pathogen infection. In manipulating the cleavage of gasdermins (GSDMs), researchers have found that GSDM proteins serve as the real executors and the deterministic players in fate decisions of pyroptotic cells. Whether inflammatory characteristics induced by pyroptosis could cause damage the host or improve immune activity is largely dependent on the context, timing, and response degree. Here, we systematically review current points involved in regulatory mechanisms and the multidimensional roles of pyroptosis in several metabolic diseases and the tumor microenvironment. Targeting pyroptosis may reveal potential therapeutic avenues.
CONFLICT OF INTEREST
The authors declare that they have no conflict of interest.
REFERENCES
- 1Zychlinsky A, Prevost MC, Sansonetti PJ. Shigella flexneri induces apoptosis in infected macrophages. Nature. 1992; 358: 167–169.
- 2Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001; 9: 113–114.
- 3Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015; 526: 660-665.
- 4Rühl S, Shkarina K, Demarco B, Heilig R, Santos JC, Broz P. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science. 2018; 362: 956–960.
- 5Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020; 21: 25–42.
- 6Zhang Z, Zhang Y, Xia S, et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature. 2020; 579: 415–420.
- 7Man SM, Karki R, Kanneganti TD. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 2017; 277: 61–75.
- 8Rosenbaum SR, Wilski NA, Aplin AE. Fueling the fire: inflammatory forms of cell death and implications for cancer immunotherapy. Cancer Discov. 2021; 11: 266–281.
- 9Tang R, Xu J, Zhang B, et al. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 2020; 13: 110.
- 10Wang Q, Wang Y, Ding J, et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature. 2020; 579: 421–426.
- 11Fang Y, Tian S, Pan Y, et al. Pyroptosis: a new frontier in cancer. Biomed Pharmacother. 2020; 121:109595.
- 12Ahechu P, Zozaya G, Martí P, et al. NLRP3 inflammasome: a possible link between obesity-associated low-grade chronic inflammation and colorectal cancer development. Front Immunol. 2018; 9: 2918.
- 13Friedlander AM. Macrophages are sensitive to anthrax lethal toxin through an acid-dependent process. J Biol Chem. 1986; 261: 7123–7126.
- 14Black RA, Kronheim SR, Merriam JE, March CJ, Hopp TP. A pre-aspartate-specific protease from human leukocytes that cleaves pro-interleukin-1 beta. J Biol Chem. 1989; 264: 5323–5326.
- 15Thornberry NA, Bull HG, Calaycay JR, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992; 356: 768–774.
- 16Cerretti DP, Kozlosky CJ, Mosley B, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992; 256: 97–100.
- 17Chen Y, Smith MR, Thirumalai K, Zychlinsky A. A bacterial invasin induces macrophage apoptosis by binding directly to ICE. EMBO J. 1996; 15: 3853–3860.
- 18Van Laer L, Huizing EH, Verstreken M, et al. Nonsyndromic hearing impairment is associated with a mutation in DFNA5. Nat Genet. 1998; 20: 194–197.
- 19Saeki N, Kuwahara Y, Sasaki H, Satoh H, Shiroishi T. Gasdermin (Gsdm) localizing to mouse chromosome 11 is predominantly expressed in upper gastrointestinal tract but significantly suppressed in human gastric cancer cells. Mamm Genome. 2000; 11: 718–724.
- 20Watabe K, Ito A, Asada H, et al. Structure, expression and chromosome mapping of MLZE, a novel gene which is preferentially expressed in metastatic melanoma cells. Japn Cancer Res. 2001; 92: 140–151.
- 21Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002; 10: 417–426.
- 22Katoh M, Katoh M. Evolutionary recombination hotspot around GSDML-GSDM locus is closely linked to the oncogenomic recombination hotspot around the PPP1R1B-ERBB2-GRB7 amplicon. Int J Oncol. 2004; 24: 757–763.
- 23Runkel F, Marquardt A, Stoeger C, et al. The dominant alopecia phenotypes Bareskin, Rex-denuded, and Reduced Coat 2 are caused by mutations in gasdermin 3. Genomics. 2004; 84: 824–835.
- 24Katoh M, Katoh M. Identification and characterization of human DFNA5L, mouse Dfna5l, and rat Dfna5l genes in silico. Int J Oncol. 2004; 25: 765–770.
- 25Delmaghani S, del Castillo FJ, Michel V, et al. Mutations in the gene encoding pejvakin, a newly identified protein of the afferent auditory pathway, cause DFNB59 auditory neuropathy. Nat Genet. 2006; 38: 770–778.
- 26Schwander M, Sczaniecka A, Grillet N, et al. A forward genetics screen in mice identifies recessive deafness traits and reveals that pejvakin is essential for outer hair cell function. J Neurosci. 2007; 27: 2163–2175.
- 27Verlaan DJ, Berlivet S, Hunninghake GM, et al. Allele-specific chromatin remodeling in the ZPBP2/GSDMB/ORMDL3 locus associated with the risk of asthma and autoimmune disease. Am J Hum Genet. 2009; 85: 377–393.
- 28Agard NJ, Maltby D, Wells JA. Inflammatory stimuli regulate caspase substrate profiles. Mol Cell Proteomics. 2010; 9: 880–893.
- 29Lei M, Bai X, Yang T, et al. Gsdma3 is a new factor needed for TNF-α-mediated apoptosis signal pathway in mouse skin keratinocytes. Histochem Cell Biol. 2012; 138: 385–396.
- 30Broz P, Ruby T, Belhocine K, et al. Caspase-11 increases susceptibility to Salmonella infection in the absence of caspase-1. Nature. 2012; 490: 288–291.
- 31Shi J, Zhao Y, Wang Y, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014; 514: 187–192.
- 32Kayagaki N, Stowe IB, Lee BL, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015; 526: 666–671.
- 33Man SM, Kanneganti TD. Gasdermin D: the long-awaited executioner of pyroptosis. Cell Res. 2015; 25: 1183–1184.
- 34Delmaghani S, Defourny J, Aghaie A, et al. Hypervulnerability to sound exposure through impaired adaptive proliferation of peroxisomes. Cell. 2015; 163: 894–906.
- 35Ding J, Wang K, Liu W, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016; 535: 111–116.
- 36Wang Y, Gao W, Shi X, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017; 547: 99–103.
- 37Rathkey JK, Zhao J, Liu Z, et al. Chemical disruption of the pyroptotic pore-forming protein gasdermin D inhibits inflammatory cell death and sepsis. Sci Immunol. 2018; 3:eaat2738.
- 38Sollberger G, Choidas A, Burn GL, et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci Immunol. 2018; 3:eaar6689.
- 39Chen KW, Monteleone M, Boucher D, et al. Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps. Sci Immunol. 2018; 3:eaar6676.
- 40Kambara H, Liu F, Zhang X, et al. Gasdermin D exerts anti-inflammatory effects by promoting neutrophil death. Cell Rep. 2018; 22: 2924–2936.
- 41Ruan J, Xia S, Liu X, Lieberman J, Wu H. Cryo-EM structure of the gasdermin A3 membrane pore. Nature. 2018; 557: 62–67.
- 42Sarhan J, Liu BC, Muendlein HI, et al. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci U S A. 2018; 115: E10888–e10897.
- 43Chen KW, Demarco B, Heilig R, et al. Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. EMBO J. 2019; 38:e101638.
- 44Burgener SS, Leborgne NGF, Snipas SJ, et al. Cathepsin G inhibition by serpinb1 and serpinb6 prevents programmed necrosis in neutrophils and monocytes and reduces GSDMD-driven inflammation. Cell Rep. 2019; 27: 3646–3656.e3645.
- 45Zhou Z, He H, Wang K, et al. Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science. 2020; 368:eaaz7548.
- 46Hou J, Zhao R, Xia W, et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol. 2020; 22: 1264–1275.
- 47Erkes DA, Cai W, Sanchez IM, et al. Mutant BRAF and MEK inhibitors regulate the tumor immune microenvironment via pyroptosis. Cancer Discov. 2020; 10: 254–269.
- 48Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, et al. Succination inactivates gasdermin D and blocks pyroptosis. Science. 2020; 369: 1633–1637.
- 49Hu JJ, Liu X, Xia S, et al. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol. 2020; 21: 736–745.
- 50Zheng M, Karki R, Vogel P, Kanneganti TD. Caspase-6 is a key regulator of innate immunity, inflammasome activation, and host defense. Cell. 2020; 181: 674–687.e613.
- 51Wang K, Sun Q, Zhong X, et al. Structural mechanism for GSDMD targeting by autoprocessed caspases in pyroptosis. Cell. 2020; 180: 941–955.e920.
- 52Xia S, Zhang Z, Magupalli VG, et al. Gasdermin D pore structure reveals preferential release of mature interleukin-1. Nature. 2021; 593: 607–611.
- 53Pentimalli F, Grelli S, Di Daniele N, Melino G, Amelio I. Cell death pathologies: targeting death pathways and the immune system for cancer therapy. Genes Immun. 2019; 20: 539–554.
- 54Hachim MY, Khalil BA, Elemam NM, Maghazachi AA. Pyroptosis: the missing puzzle among innate and adaptive immunity crosstalk. J Leukoc Biol. 2020; 108: 323–338.
- 55He WT, Wan H, Hu L, et al. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 2015; 25: 1285–1298.
- 56Kayagaki N, Warming S, Lamkanfi M, et al. Non-canonical inflammasome activation targets caspase-11. Nature. 2011; 479: 117–121.
- 57Schmid-Burgk JL, Gaidt MM, Schmidt T, Ebert TS, Bartok E, Hornung V. Caspase-4 mediates non-canonical activation of the NLRP3 inflammasome in human myeloid cells. Eur J Immunol. 2015; 45: 2911–2917.
- 58Baker PJ, Boucher D, Bierschenk D, et al. NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur J Immunol. 2015; 45: 2918–2926.
- 59Ruan J, Wang S, Wang J. Mechanism and regulation of pyroptosis-mediated in cancer cell death. Chem Biol Interact. 2020; 323:109052.
- 60Liu X, Zhang Z, Ruan J, et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 2016; 535: 153–158.
- 61Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014; 157: 1013–1022.
- 62Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016; 16: 407–420.
- 63Sollberger G, Strittmatter GE, Garstkiewicz M, Sand J, Beer HD. Caspase-1: the inflammasome and beyond. Innate Immunity 2014; 20: 115–125.
- 64Yin Q, Fu TM, Li J, Wu H. Structural biology of innate immunity. Annu Rev Immunol. 2015; 33: 393–416.
- 65Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140: 821–832.
- 66Chen X, He WT, Hu L, et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis. Cell Res. 2016; 26: 1007–1020.
- 67Sborgi L, Rühl S, Mulvihill E, et al. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO J. 2016; 35: 1766–1778.
- 68Kuriakose T, Kanneganti TD. Gasdermin D flashes an exit signal for IL-1. Immunity. 2018; 48: 1–3.
- 69Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019; 19: 477–489.
- 70Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019; 20: 3328.
- 71Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011; 469: 221–225.
- 72Shirasuna K, Karasawa T, Takahashi M. Exogenous nanoparticles and endogenous crystalline molecules as danger signals for the NLRP3 inflammasomes. J Cell Physiol. 2019; 234: 5436–5450.
- 73He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016; 41: 1012–1021.
- 74Chavarría-Smith J, Vance RE. Direct proteolytic cleavage of NLRP1B is necessary and sufficient for inflammasome activation by anthrax lethal factor. PLoS Pathog. 2013; 9:e1003452.
- 75Levinsohn JL, Newman ZL, Hellmich KA, et al. Anthrax lethal factor cleavage of Nlrp1 is required for activation of the inflammasome. PLoS Pathog. 2012; 8:e1002638.
- 76Ewald SE, Chavarria-Smith J, Boothroyd JC. NLRP1 is an inflammasome sensor for Toxoplasma gondii. Infect Immun. 2014; 82: 460–468.
- 77Franchi L, Amer A, Body-Malapel M, et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1beta in salmonella-infected macrophages. Nat Immunol. 2006; 7: 576–582.
- 78Lightfield KL, Persson J, Brubaker SW, et al. Critical function for Naip5 in inflammasome activation by a conserved carboxy-terminal domain of flagellin. Nat Immunol. 2008; 9: 1171–1178.
- 79Kofoed EM, Vance RE. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity. Nature. 2011; 477: 592–595.
- 80Zhao Y, Yang J, Shi J, et al. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature. 2011; 477: 596–600.
- 81Zhang L, Chen S, Ruan J, et al. Cryo-EM structure of the activated NAIP2-NLRC4 inflammasome reveals nucleated polymerization. Science. 2015; 350: 404–409.
- 82Duncan JA, Canna SW. The NLRC4 inflammasome. Immunol Rev. 2018; 281: 115–123.
- 83Hornung V, Ablasser A, Charrel-Dennis M, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009; 458: 514–518.
- 84Roberts TL, Idris A, Dunn JA, et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science. 2009; 323: 1057–1060.
- 85Rathinam VA, Jiang Z, Waggoner SN, et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol. 2010; 11: 395–402.
- 86Jin T, Perry A, Jiang J, et al. Structures of the HIN domain:dNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity. 2012; 36: 561–571.
- 87Matyszewski M, Morrone SR, Sohn J. Digital signaling network drives the assembly of the AIM2-ASC inflammasome. Proc Natl Acad Sci U S A. 2018; 115: E1963–e1972.
- 88Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012; 13: 333–342.
- 89Xu H, Yang J, Gao W, et al. Innate immune sensing of bacterial modifications of Rho GTPases by the Pyrin inflammasome. Nature. 2014; 513: 237–241.
- 90Chung LK, Park YH, Zheng Y, et al. The Yersinia virulence factor YopM hijacks host kinases to inhibit type III effector-triggered activation of the pyrin inflammasome. Cell Host Microbe. 2016; 20: 296–306.
- 91Park YH, Wood G, Kastner DL, Chae JJ. Pyrin inflammasome activation and RhoA signaling in the autoinflammatory diseases FMF and HIDS. Nat Immunol. 2016; 17: 914–921.
- 92Gao W, Yang J, Liu W, Wang Y, Shao F. Site-specific phosphorylation and microtubule dynamics control pyrin inflammasome activation. Proc Natl Acad Sci U S A. 2016; 113: E4857–4866.
- 93Aglietti RA, Estevez A, Gupta A, et al. GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc Natl Acad Sci U S A. 2016; 113: 7858–7863.
- 94Yang D, He Y, Muñoz-Planillo R, Liu Q, Núñez G. Caspase-11 requires the pannexin-1 channel and the purinergic P2X7 pore to mediate pyroptosis and endotoxic shock. Immunity. 2015; 43: 923–932.
- 95Rühl S, Broz P. Caspase-11 activates a canonical NLRP3 inflammasome by promoting K(+) efflux. Eur J Immunol. 2015; 45: 2927–2936.
- 96Shi J, Gao W, Shao F. Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem Sci. 2017; 42: 245–254.
- 97Rogers C, Erkes DA, Nardone A, Aplin AE, Fernandes-Alnemri T, Alnemri ES. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat Commun. 2019; 10: 1689.
- 98Huang LS, Hong Z, Wu W, et al. mtDNA activates cGAS signaling and suppresses the YAP-mediated endothelial cell proliferation program to promote inflammatory injury. Immunity. 2020; 52: 475–486.e475.
- 99Schwarzer R, Jiao H, Wachsmuth L, Tresch A. Pasparakis M FADD and caspase-8 regulate gut homeostasis and inflammation by controlling MLKL- and GSDMD-mediated death of intestinal epithelial cells. Immunity. 2020; 52: 978–993.e976.
- 100Orning P, Weng D, Starheim K, et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science. 2018; 362: 1064–1069.
- 101Newton K, Wickliffe KE, Maltzman A, et al. Activity of caspase-8 determines plasticity between cell death pathways. Nature. 2019; 575: 679–682.
- 102Muendlein HI, Jetton D, Connolly WM, et al. cFLIP(L) protects macrophages from LPS-induced pyroptosis via inhibition of complex II formation. Science. 2020; 367: 1379–1384.
- 103Zhou B, Zhang JY, Liu XS, et al. Tom20 senses iron-activated ROS signaling to promote melanoma cell pyroptosis. Cell Res. 2018; 28: 1171–1185.
- 104de Vasconcelos NM, Van Opdenbosch N, Van Gorp H, et al. An apoptotic caspase network safeguards cell death induction in pyroptotic macrophages. Cell Rep. 2020; 32:107959.
- 105Li L, Li Y, Bai Y. Role of GSDMB in pyroptosis and cancer. Cancer Manag Res. 2020; 12: 3033–3043.
- 106Chao KL, Kulakova L, Herzberg O. Gene polymorphism linked to increased asthma and IBD risk alters gasdermin-B structure, a sulfatide and phosphoinositide binding protein. Proc Natl Acad Sci U S A. 2017; 114: E1128–e1137.
- 107Evavold CL, Ruan J, Tan Y, Xia S, Wu H, Kagan JC, The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages. Immunity. 2018; 48: 35–44.e36.
- 108Madore AM, Pain L, Boucher-Lafleur AM, et al. Asthma-associated polymorphisms in 17q12-21 locus modulate methylation and gene expression of GSDMA in naïve CD4(+) T cells. J Genet Genomics. 2020; 47: 171–174.
- 109Moreno-Moral A, Bagnati M, Koturan S, et al. Changes in macrophage transcriptome associate with systemic sclerosis and mediate GSDMA contribution to disease risk. Ann Rheum Dis. 2018; 77: 596–601.
- 110Lin PH, Lin HY, Kuo CC, Yang LT. N-terminal functional domain of Gasdermin A3 regulates mitochondrial homeostasis via mitochondrial targeting. J Biomed Sci. 2015; 22: 44.
- 111Saeki N, Kim DH, Usui T, et al. GASDERMIN, suppressed frequently in gastric cancer, is a target of LMO1 in TGF-beta-dependent apoptotic signalling. Oncogene. 2007; 26: 6488–6498.
- 112Li X, Christenson SA, Modena B, et al. Genetic analyses identify GSDMB associated with asthma severity, exacerbations, and antiviral pathways. J Allergy Clin Immunol. 2021; 147: 894–909.
- 113Saleh NM, Raj SM, Smyth DJ, et al. Genetic association analyses of atopic illness and proinflammatory cytokine genes with type 1 diabetes. Diabetes Metab Res Rev. 2011; 27: 838–843.
- 114Kaur S, Mirza AH, Overgaard AJ, Pociot F, Størling J, A dual systems genetics approach identifies common genes, networks, and pathways for type 1 and 2 diabetes in human islets. Front Genet. 2021; 12:630109.
- 115Li T, Ortiz-Fernández L, Andrés-León E, et al. Epigenomics and transcriptomics of systemic sclerosis CD4+ T cells reveal long-range dysregulation of key inflammatory pathways mediated by disease-associated susceptibility loci. Genome Med. 2020; 12: 81.
- 116Gui H, Levin AM, Hu D, et al. Mapping the 17q12-21.1 locus for variants associated with early-onset asthma in African Americans. Am J Respir Crit Care Med. 2021; 203: 424–436.
10.1164/rccm.202006-2623OC Google Scholar
- 117Liu D, Zhou D, Sun Y, et al. A transcriptome-wide association study identifies candidate susceptibility genes for pancreatic cancer risk. Cancer Res. 2020; 80: 4346–4354.
- 118Molina-Crespo Á, Cadete A, Sarrio D, et al. Intracellular delivery of an antibody targeting gasdermin-B reduces HER2 breast cancer aggressiveness. Clin Cancer Res. 2019; 25: 4846–4858.
- 119Hu Y, Jin S, Cheng L, Liu G, Jiang Q. Autoimmune disease variants regulate GSDMB gene expression in human immune cells and whole blood. Proc Natl Acad Sci U S A.. 2017; 114: E7860–E7862.
- 120Calışkan M, Bochkov YA, Kreiner-Møller E, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. N Engl J Med. 2013; 368: 1398–1407.
- 121Christodoulou K, Wiskin AE, Gibson J, et al. Next generation exome sequencing of paediatric inflammatory bowel disease patients identifies rare and novel variants in candidate genes. Gut. 2013; 62: 977–984.
- 122Jiang H, Moro A, Liu Y, et al. Two GWAS-identified variants are associated with lumbar spinal stenosis and Gasdermin-C expression in Chinese population. Sci Rep. 2020; 10:21069.
- 123Shou Y, Yang L, Yang Y, Zhu X, Li F, Xu J. Identification of signatures of prognosis prediction for melanoma using a hypoxia score. Front Genet. 2020; 11:570530.
- 124Saeki N, Usui T, Aoyagi K, et al. Distinctive expression and function of four GSDM family genes (GSDMA-D) in normal and malignant upper gastrointestinal epithelium. Genes Chromosomes Cancer. 2009; 48: 261–271.
- 125Miguchi M, Hinoi T, Shimomura M, et al. Gasdermin C is upregulated by inactivation of transforming growth factor β receptor type II in the presence of mutated apc, promoting colorectal cancer proliferation. PLoS One. 2016; 11:e0166422.
- 126Tamura M, Tanaka S, Fujii T, et al. Members of a novel gene family, Gsdm, are expressed exclusively in the epithelium of the skin and gastrointestinal tract in a highly tissue-specific manner. Genomics. 2007; 89: 618–629.
- 127Russo AJ, Vasudevan SO, Méndez-Huergo SP, et al. Intracellular immune sensing promotes inflammation via gasdermin D-driven release of a lectin alarmin. Nat Immunol. 2021; 22: 154–165.
- 128Kiss M, Vande Walle L, Saavedra PHV, et al. IL1β promotes immune suppression in the tumor microenvironment independent of the inflammasome and gasdermin D. Cancer Immunol Res. 2021; 9: 309–323.
10.1158/2326-6066.CIR-20-0431 Google Scholar
- 129Gaul S, Leszczynska A, Alegre F, et al. Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis. J Hepatol. 2021; 74: 156–167.
- 130Zhang H, Zeng L, Xie M, et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe. 2020; 27: 556–570.e556.
- 131Yang M, So KF, Lo ACY, Lam WC. The effect of lycium barbarum polysaccharides on pyroptosis-associated amyloid β(1-40) oligomers-induced adult retinal pigment epithelium 19 cell damage. Int J Mol Sci. 2020; 21: 4658.
10.3390/ijms21134658 Google Scholar
- 132Li S, Wu Y, Yang D, et al. Gasdermin D in peripheral myeloid cells drives neuroinflammation in experimental autoimmune encephalomyelitis. J Exp Med. 2019; 216: 2562–2581.
- 133Xiao J, Wang C, Yao JC, et al. Gasdermin D mediates the pathogenesis of neonatal-onset multisystem inflammatory disease in mice. PLoS Biol. 2018; 16:e3000047.
- 134Xia W, Li Y, Wu M, et al. Gasdermin E deficiency attenuates acute kidney injury by inhibiting pyroptosis and inflammation. Cell Death Dis. 2021; 12: 139.
- 135Li Y, Yuan Y, Huang ZX, et al. GSDME-mediated pyroptosis promotes inflammation and fibrosis in obstructive nephropathy. Cell Death Differ. 2021. https://doi.org/10.1038/s41418-021-00755-6.
- 136An H, Heo JS, Kim P, et al. Tetraarsenic hexoxide enhances generation of mitochondrial ROS to promote pyroptosis by inducing the activation of caspase-3/GSDME in triple-negative breast cancer cells. Cell Death Dis. 2021; 12: 159.
- 137Shen X, Wang H, Weng C, Jiang H, Chen J. Caspase 3/GSDME-dependent pyroptosis contributes to chemotherapy drug-induced nephrotoxicity. Cell Death Dis. 2021; 12: 186.
- 138Ibrahim J, De Schutter E, Op de Beeck K. GSDME: a potential ally in cancer detection and treatment. Trends Cancer. 2021; 7: 392–394.
10.1016/j.trecan.2020.12.002 Google Scholar
- 139Tan G, Huang C, Chen J, Zhi F. HMGB1 released from GSDME-mediated pyroptotic epithelial cells participates in the tumorigenesis of colitis-associated colorectal cancer through the ERK1/2 pathway. J Hematol Oncol. 2020; 13: 149.
- 140Kazmierczak M, Kazmierczak P, Peng AW, et al. Pejvakin, a candidate stereociliary rootlet protein, regulates hair cell function in a cell-autonomous manner. J Neurosci. 2017; 37: 3447–3464.
- 141Collin RW, Kalay E, Oostrik J, et al. Involvement of DFNB59 mutations in autosomal recessive nonsyndromic hearing impairment. Hum Mutat. 2007; 28: 718–723.
- 142Hao K, Bossé Y, Nickle DC, et al. Lung eQTLs to help reveal the molecular underpinnings of asthma. PLos Genet. 2012; 8:e1003029.
- 143Lunny DP, Weed E, Nolan PM, Marquardt A, Augustin M, Porter RM. Mutations in gasdermin 3 cause aberrant differentiation of the hair follicle and sebaceous gland. J Invest Dermatol. 2005; 124: 615–621.
- 144Yu J, Kang MJ, Kim BJ, et al. Polymorphisms in GSDMA and GSDMB are associated with asthma susceptibility, atopy and BHR. Pediatr Pulmonol. 2011; 46: 701–708.
- 145Kumar S, Rathkolb B, Budde BS, et al. Gsdma3(I359N) is a novel ENU-induced mutant mouse line for studying the function of Gasdermin A3 in the hair follicle and epidermis. J Dermatol Sci. 2012; 67: 190–192.
- 146Tamura M, Shiroishi T. GSDM family genes meet autophagy. Biochem J. 2015; 469: e5–e7.
- 147Shi P, Tang A, Xian L, et al. Loss of conserved Gsdma3 self-regulation causes autophagy and cell death. Biochem J. 2015; 468: 325–336.
- 148Carl-McGrath S, Schneider-Stock R, Ebert M, Röcken C. Differential expression and localisation of gasdermin-like (GSDML), a novel member of the cancer-associated GSDMDC protein family, in neoplastic and non-neoplastic gastric, hepatic, and colon tissues. Pathology. 2008; 40: 13–24.
- 149Panganiban RA, Sun M, Dahlin A, et al. A functional splice variant associated with decreased asthma risk abolishes the ability of gasdermin B to induce epithelial cell pyroptosis. J Allergy Clin Immunol. 2018; 142: 1469–1478.e1462.
- 150Clark AD, Nair N, Anderson AE, et al. Lymphocyte DNA methylation mediates genetic risk at shared immune-mediated disease loci. J Allergy Clin Immunol. 2020; 145: 1438–1451.
10.1016/j.jaci.2019.12.910 Google Scholar
- 151Sun Q, Yang J, Xing G, Sun Q, Zhang L, He F. Expression of GSDML associates with tumor progression in uterine cervix cancer. Transl Oncol. 2008; 1: 73–83.
- 152Hergueta-Redondo M, Sarrio D, Molina-Crespo Á, et al. Gasdermin B expression predicts poor clinical outcome in HER2-positive breast cancer. Oncotarget. 2016; 7: 56295–56308.
- 153Hergueta-Redondo M, Sarrió D, Molina-Crespo Á, et al. Gasdermin-B promotes invasion and metastasis in breast cancer cells. PLoS One. 2014; 9:e90099.
- 154Ntontsi P, Photiades A, Zervas E, Xanthou G, Samitas K. Genetics and epigenetics in asthma. Int J Mol Sci. 2021; 22: 2412.
- 155Wei J, Xu Z, Chen X, et al. Overexpression of GSDMC is a prognostic factor for predicting a poor outcome in lung adenocarcinoma. Mol Med Rep. 2020; 21: 360–370.
- 156Kusumaningrum N, Lee DH, Yoon HS, Park CH, Chung JH. Ultraviolet light-induced gasdermin C expression is mediated via TRPV1/calcium/calcineurin/NFATc1 signaling. Int J Mol Med. 2018; 42: 2859–2866.
- 157Kusumaningrum N, Lee DH, Yoon HS, Kim YK, Park CH, Chung JH. Gasdermin C is induced by ultraviolet light and contributes to MMP-1 expression via activation of ERK and JNK pathways. J Dermatol Sci. 2018; 90: 180–189.
- 158Suri P, Palmer MR, Tsepilov YA, et al. Genome-wide meta-analysis of 158,000 individuals of European ancestry identifies three loci associated with chronic back pain. PLos Genet. 2018; 14:e1007601.
- 159Blanc F, Maroilley T, Revilla M, et al. Influence of genetics and the pre-vaccination blood transcriptome on the variability of antibody levels after vaccination against Mycoplasma hyopneumoniae in pigs. Genet Sel Evol. 2021; 53: 24.
- 160Zhang Y, Fukui N, Yahata M, et al. Genome-wide DNA methylation profile implicates potential cartilage regeneration at the late stage of knee osteoarthritis. Osteoarthritis Cartilage. 2016; 24: 835–843.
- 161Zhang Y, Fukui N, Yahata M, et al. Identification of DNA methylation changes associated with disease progression in subchondral bone with site-matched cartilage in knee osteoarthritis. Sci Rep. 2016; 6:34460.
- 162Bjornsdottir G, Benonisdottir S, Sveinbjornsson G, et al. Sequence variant at 8q24.21 associates with sciatica caused by lumbar disc herniation. Nat Commun. 2017; 8:14265.
- 163Crosslin DR, McDavid A, Weston N, et al. Genetic variation associated with circulating monocyte count in the eMERGE Network. Hum Mol Genet. 2013; 22: 2119–2127.
- 164Xia X, Wang X, Cheng Z, et al. The role of pyroptosis in cancer: pro-cancer or pro-“host”? Cell Death Dis. 2019; 10: 650.
- 165Du M, Yuan T, Schilter KF, et al. Prostate cancer risk locus at 8q24 as a regulatory hub by physical interactions with multiple genomic loci across the genome. Hum Mol Genet. 2015; 24: 154–166.
- 166Tseng YY, Moriarity BS, Gong W, et al. PVT1 dependence in cancer with MYC copy-number increase. Nature. 2014; 512: 82–86.
- 167Liu X, Lieberman J. Knocking 'em dead: pore-forming proteins in immune defense. Annu Rev Immunol. 2020; 38: 455–485.
- 168Fujii T, Tamura M, Tanaka S, et al. Gasdermin D (Gsdmd) is dispensable for mouse intestinal epithelium development. Genesis. 2008; 46: 418–423.
- 169Kayagaki N, Lee BL, Stowe IB, et al. IRF2 transcriptionally induces GSDMD expression for pyroptosis. Sci Signal. 2019; 12:eaax4917.
- 170Liao W, Overman MJ, Boutin AT, et al. KRAS-IRF2 axis drives immune suppression and immune therapy resistance in colorectal cancer. Cancer Cell. 2019; 35: 559–572.e557.
- 171Han C, Yang Y, Yu A, et al. Investigation on the mechanism of mafenide in inhibiting pyroptosis and the release of inflammatory factors. Eur J Pharm Sci. 2020; 147:105303.
10.1016/j.ejps.2020.105303 Google Scholar
- 172Qiu X, Cheng X, Zhang J, Yuan C, Zhao M, Yang X. Ethyl pyruvate confers protection against endotoxemia and sepsis by inhibiting caspase-11-dependent cell pyroptosis. Int Immunopharmacol. 2020; 78:106016.
10.1016/j.intimp.2019.106016 Google Scholar
- 173Kanneganti A, Malireddi RKS, Saavedra PHV, et al. GSDMD is critical for autoinflammatory pathology in a mouse model of Familial Mediterranean Fever. J Exp Med. 2018; 215: 1519–1529.
- 174De Schutter E, Croes L, Ibrahim J, et al. GSDME and its role in cancer: from behind the scenes to the front of the stage. Int J Cancer. 2021; 148: 2872–2883.
- 175de Beeck KO, Van Laer L, Van Camp G. DFNA5, a gene involved in hearing loss and cancer: a review. Ann Otol Rhinol Laryngol. 2012; 121: 197–207.
- 176Masuda Y, Futamura M, Kamino H, et al. The potential role of DFNA5, a hearing impairment gene, in p53-mediated cellular response to DNA damage. J Hum Genet. 2006; 51: 652–664.
- 177Rogers C, Fernandes-Alnemri T, Mayes L, Alnemri D, Cingolani G, Alnemri ES. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat Commun. 2017; 8:14128.
- 178Hu L, Chen M, Chen X, et al. Chemotherapy-induced pyroptosis is mediated by BAK/BAX-caspase-3-GSDME pathway and inhibited by 2-bromopalmitate. Cell Death Dis. 2020; 11: 281.
- 179Salime S, Charif M, Bousfiha A, et al. Homozygous mutations in PJVK and MYO15A genes associated with non-syndromic hearing loss in Moroccan families. Int J Pediatr Otorhinolaryngol. 2017; 101: 25–29.
10.1016/j.ijporl.2017.07.024 Google Scholar
- 180Defourny J, Aghaie A, Perfettini I, Avan P, Delmaghani S, Petit C. Pejvakin-mediated pexophagy protects auditory hair cells against noise-induced damage. Proc Natl Acad Sci U S A. 2019; 116: 8010–8017.
- 181Mujtaba G, Bukhari I, Fatima A, Naz S. A p.C343S missense mutation in PJVK causes progressive hearing loss. Gene. 2012; 504: 98–101.
10.1016/j.gene.2012.05.013 Google Scholar
- 182Cheng YF, Tsai YH, Huang CY, et al. Generation and pathological characterization of a transgenic mouse model carrying a missense PJVK mutation. Biochem Biophys Res Commun. 2020; 532: 675–681.
10.1016/j.bbrc.2020.07.101 Google Scholar
- 183Harris SL, Kazmierczak M, Pangršič T, et al. Conditional deletion of pejvakin in adult outer hair cells causes progressive hearing loss in mice. Neuroscience. 2017; 344: 380–393.
- 184Zhou Y, Tariq M, He S, Abdullah U, Zhang J, Baig SM. Whole exome sequencing identified mutations causing hearing loss in five consanguineous Pakistani families. BMC Med Genet. 2020; 21: 151.
- 185Khan A, Han S, Wang R, Ansar M, Ahmad W, Zhang X. Sequence variants in genes causing nonsyndromic hearing loss in a Pakistani cohort. Mol Genet Genomic Med. 2019; 7:e917.
- 186Carvalho GM, Ramos PZ, Castilho AM, Guimarães AC, Sartorato EL. Molecular study of patients with auditory neuropathy. Mol Med Rep. 2016; 14: 481–490.
- 187Borck G, Rainshtein L, Hellman-Aharony S, et al. High frequency of autosomal-recessive DFNB59 hearing loss in an isolated Arab population in Israel. Clin Genet. 2012; 82: 271–276.
- 188Wu CC, Lin YH, Liu TC, et al. Identifying children with poor cochlear implantation outcomes using massively parallel sequencing. Medicine (Baltimore). 2015; 94:e1073.
- 189Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006; 440: 237–241.
- 190Renaudin F, Orliaguet L, Castelli F, et al. Gout and pseudo-gout-related crystals promote GLUT1-mediated glycolysis that governs NLRP3 and interleukin-1β activation on macrophages. Ann Rheum Dis. 2020; 79: 1506–1514.
- 191Hao K, Jiang W, Zhou M, et al. Targeting BRD4 prevents acute gouty arthritis by regulating pyroptosis. Int J Biol Sci. 2020; 16: 3163–3173.
- 192He X, Fan X, Bai B, Lu N, Zhang S, Zhang L, Pyroptosis is a critical immune-inflammatory response involved in atherosclerosis. Pharmacol Res. 2021; 165:105447.
- 193Yang M, Lv H, Liu Q, et al. Colchicine alleviates cholesterol crystal-induced endothelial cell pyroptosis through activating AMPK/SIRT1 pathway. Oxid Med Cell Longev. 2020; 2020:9173530.
- 194Chen JJ, Tao J, Zhang XL, et al. Inhibition of the ox-LDL-induced pyroptosis by FGF21 of human umbilical vein endothelial cells through the TET2-UQCRC1-ROS pathway. DNA Cell Biol. 2020; 39: 661–670.
- 195Zhaolin Z, Jiaojiao C, Peng W, et al. OxLDL induces vascular endothelial cell pyroptosis through miR-125a-5p/TET2 pathway. J Cell Physiol. 2019; 234: 7475–7491.
- 196Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J, Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010; 11: 136–140.
- 197Reynolds CM, McGillicuddy FC, Harford KA, Finucane OM, Mills KH, Roche HM. Dietary saturated fatty acids prime the NLRP3 inflammasome via TLR4 in dendritic cells-implications for diet-induced insulin resistance. Mol Nutr Food Res. 2012; 56: 1212–1222.
- 198Rojas J, Bermudez V, Palmar J, et al. Pancreatic beta cell death: novel potential mechanisms in diabetes therapy. J Diabetes Res. 2018; 2018:9601801.
- 199Gu J, Huang W, Zhang W, et al. Sodium butyrate alleviates high-glucose-induced renal glomerular endothelial cells damage via inhibiting pyroptosis. Int Immunopharmacol. 2019; 75:105832.
- 200Gubser PM, Bantug GR, Razik L, et al. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat Immunol. 2013; 14: 1064–1072.
- 201Fox CJ, Hammerman PS, Thompson CB. Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol. 2005; 5: 844–852.
- 202Macintyre AN, Gerriets VA, Nichols AG, et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 2014; 20: 61–72.
- 203Freemerman AJ, Johnson AR, Sacks GN, et al. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J Biol Chem. 2014; 289: 7884–7896.
- 204Tannahill GM, Curtis AM, Adamik J, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013; 496: 238–242.
- 205Dror E, Dalmas E, Meier DT, et al. Postprandial macrophage-derived IL-1β stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol. 2017; 18: 283–292.
- 206Takahashi T, Suzuki T. Role of sulfatide in normal and pathological cells and tissues. J Lipid Res. 2012; 53: 1437–1450.
- 207Ricchi M, Odoardi MR, Carulli L, et al. Differential effect of oleic and palmitic acid on lipid accumulation and apoptosis in cultured hepatocytes. J Gastroenterol Hepatol. 2009; 24: 830–840.
- 208Cnop M, Hannaert JC, Hoorens A, Eizirik DL, Pipeleers DG. Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation. Diabetes. 2001; 50: 1771–1777.
- 209Zhan ZY, Wu M, Shang Y, et al. Taxifolin ameliorate high-fat-diet feeding plus acute ethanol binge-induced steatohepatitis through inhibiting inflammatory caspase-1-dependent pyroptosis. Food Funct. 2021; 12: 362–372.
10.1039/D0FO02653K Google Scholar
- 210Zeng X, Zhu M, Liu X, et al. Oleic acid ameliorates palmitic acid induced hepatocellular lipotoxicity by inhibition of ER stress and pyroptosis. Nutr Metab. 2020; 17: 11.
- 211Koh EH, Yoon JE, Ko MS, et al. Sphingomyelin synthase 1 mediates hepatocyte pyroptosis to trigger non-alcoholic steatohepatitis. Gut. 2020. http://doi.org/10.1136/gutjnl-2020-322509.
10.1136/gutjnl-2020-322509 Google Scholar
- 212Yabuta S, Shidoji Y. TLR4-mediated pyroptosis in human hepatoma-derived HuH-7 cells induced by a branched-chain polyunsaturated fatty acid, geranylgeranoic acid. Biosci Rep. 2020; 40:BSR20194118.
- 213Zhaolin Z, Guohua L, Shiyuan W, Zuo W. Role of pyroptosis in cardiovascular disease. Cell Prolif. 2019; 52:e12563.
- 214Yin Y, Li X, Sha X, et al. Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arterioscler Thromb Vasc Biol. 2015; 35: 804–816.
- 215Zeng Z, Zheng Q, Chen J, et al. FGF21 mitigates atherosclerosis via inhibition of NLRP3 inflammasome-mediated vascular endothelial cells pyroptosis. Exp Cell Res. 2020; 393:112108.
- 216Li Y, Niu X, Xu H, et al. VX-765 attenuates atherosclerosis in ApoE deficient mice by modulating VSMCs pyroptosis. Exp Cell Res. 2020; 389:111847.
- 217Hu Q, Zhang T, Yi L, Zhou X, Mi M, Dihydromyricetin inhibits NLRP3 inflammasome-dependent pyroptosis by activating the Nrf2 signaling pathway in vascular endothelial cells. Biofactors. 2018; 44: 123–136.
- 218Lopez-Pastrana J, Ferrer LM, Li YF, et al. Inhibition of caspase-1 activation in endothelial cells improves angiogenesis: a novel therapeutic potential for ischemia. J Biol Chem. 2015; 290: 17485–17494.
- 219An N, Gao Y, Si Z, et al. Regulatory mechanisms of the NLRP3 inflammasome, a novel immune-inflammatory marker in cardiovascular diseases. Front Immunol. 2019; 10: 1592.
- 220Ozaki E, Campbell M, Doyle SL. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015; 8: 15–27.
- 221Xiao H, Lu M, Lin TY, et al. Sterol regulatory element binding protein 2 activation of NLRP3 inflammasome in endothelium mediates hemodynamic-induced atherosclerosis susceptibility. Circulation. 2013; 128: 632–642.
- 222Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010; 464: 1357–1361.
- 223Liu Y, Tie L. Apolipoprotein M and sphingosine-1-phosphate complex alleviates TNF-α-induced endothelial cell injury and inflammation through PI3K/AKT signaling pathway. BMC Cardiovasc Disord. 2019; 19: 279.
- 224Meng Q, Li Y, Ji T, et al. Estrogen prevent atherosclerosis by attenuating endothelial cell pyroptosis via activation of estrogen receptor α-mediated autophagy. J Adv Res. 2021; 28: 149–164.
- 225Wu P, Chen J, Chen J, et al. Trimethylamine N-oxide promotes apoE(-/-) mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. J Cell Physiol. 2020; 235: 6582–6591.
- 226McCully KS. Homocysteine metabolism, atherosclerosis, and diseases of aging. Compr Physiol. 2015; 6: 471–505.
- 227Geovanini GR, Libby P. Atherosclerosis and inflammation: overview and updates. Clin Sci (Lond). 2018; 132: 1243–1252.
- 228Xi H, Zhang Y, Xu Y, et al. Caspase-1 inflammasome activation mediates homocysteine-induced pyrop-apoptosis in endothelial cells. Circ Res. 2016; 118: 1525–1539.
- 229Corrêa R, Silva LFF, Ribeiro DJS, et al. Lysophosphatidylcholine induces NLRP3 inflammasome-mediated foam cell formation and pyroptosis in human monocytes and endothelial cells. Front Immunol. 2019; 10: 2927.
- 230Li X, Du N, Zhang Q, et al. MicroRNA-30d regulates cardiomyocyte pyroptosis by directly targeting foxo3a in diabetic cardiomyopathy. Cell Death Dis. 2014; 5:e1479.
- 231Stienstra R, Joosten LA, Koenen T, et al. The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. Cell Metab. 2010; 12: 593–605.
- 232Liu J, Wang Y, Meng H, et al. Butyrate rather than LPS subverts gingival epithelial homeostasis by downregulation of intercellular junctions and triggering pyroptosis. J Clin Periodontol. 2019; 46: 894–907.
- 233Li A, Zhang S, Li J, Liu K, Huang F, Liu B, Metformin and resveratrol inhibit Drp1-mediated mitochondrial fission and prevent ER stress-associated NLRP3 inflammasome activation in the adipose tissue of diabetic mice. Mol Cell Endocrinol. 2016; 434: 36–47.
- 234Wang Y, Zhu X, Yuan S, et al. TLR4/NF-κB signaling induces GSDMD-related pyroptosis in tubular cells in diabetic kidney disease. Front Endocrinol. 2019; 10: 603.
- 235Zhu B, Cheng X, Jiang Y, et al. Silencing of KCNQ1OT1 decreases oxidative stress and pyroptosis of renal tubular epithelial cells. Diabetes Metab Syndr Obes. 2020; 13: 365–375.
- 236Xie C, Guo ZX, Qiu T, Tuo X. lncRNA GAS5/miR-452-5p reduces oxidative stress and pyroptosis of high-glucose-stimulated renal tubular cells. Diabetes Metab Syndr Obes. 2019; 12: 2609–2617.
- 237Wei M, Li Z, Xiao L, Yang Z. Effects of ROS-relative NF-κB signaling on high glucose-induced TLR4 and MCP-1 expression in podocyte injury. Mol Immunol. 2015; 68: 261–271.
- 238Yang S, Zhang J, Wang S, Zhao X, Shi J. SOCS2 overexpression alleviates diabetic nephropathy in rats by inhibiting the TLR4/NF-κB pathway. Oncotarget. 2017; 8: 91185–91198.
10.18632/oncotarget.20434 Google Scholar
- 239Aki T, Funakoshi T, Uemura K. Regulated necrosis and its implications in toxicology. Toxicology. 2015; 333: 118–126.
- 240Vandanmagsar B, Youm YH, Ravussin A, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011; 17: 179–188.
- 241Horng T, Hotamisligil GS. Linking the inflammasome to obesity-related disease. Nat Med. 2011; 17: 164–165.
- 242Qian S, Pan J, Su Y, et al. BMPR2 promotes fatty acid oxidation and protects white adipocytes from cell death in mice. Commun Biol. 2020; 3: 200.
- 243Giordano A, Murano I, Mondini E, et al. Obese adipocytes show ultrastructural features of stressed cells and die of pyroptosis. J Lipid Res. 2013; 54: 2423–2436.
- 244Pillon NJ, Chan KL, Zhang S, et al. Saturated fatty acids activate caspase-4/5 in human monocytes, triggering IL-1β and IL-18 release. Am J Physiol Endocrinol Metab. 2016; 311: E825–e835.
- 245Wang Y, Li C, Ali I, Li L, Wang G. N-acetylcysteine modulates non-esterified fatty acid-induced pyroptosis and inflammation in granulosa cells. Mol Immunol. 2020; 127: 157–163.
- 246Pizato N, Luzete BC, Kiffer L, et al. Omega-3 docosahexaenoic acid induces pyroptosis cell death in triple-negative breast cancer cells. Sci Rep. 2018; 8: 1952.
- 247Zasłona Z, Flis E, Wilk MM, et al. Caspase-11 promotes allergic airway inflammation. Nat Commun. 2020; 11: 1055.
- 248Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017; 17: 97–111.
- 249Ben-Sasson SZ, Hogg A, Hu-Li J, et al. IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells. J Exp Med. 2013; 210: 491–502.
- 250Jain A, Song R, Wakeland EK, Pasare C. T cell-intrinsic IL-1R signaling licenses effector cytokine production by memory CD4 T cells. Nat Commun. 2018; 9: 3185.
- 251Bent R, Moll L, Grabbe S, Bros M. Interleukin-1 beta-A friend or foe in malignancies? Int J Mol Sci. 2018; 19: 2155.
- 252Dinarello CA. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev. 2018; 281: 8–27.
- 253Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol. 2014; 26: 38–47.
- 254Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014; 6:a016295.
- 255Mariathasan S, Weiss DS, Newton K, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006; 440: 228–232.
- 256Wang Q, Imamura R, Motani K, Kushiyama H, Nagata S, Suda T. Pyroptotic cells externalize eat-me and release find-me signals and are efficiently engulfed by macrophages. Int Immunol. 2013; 25: 363–372.
- 257Yu M, Wang H, Ding A, et al. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock. 2006; 26: 174–179.
- 258Apetoh L, Ghiringhelli F, Tesniere A, et al. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev. 2007; 220: 47–59.
- 259Werfel TA, Cook RS. Efferocytosis in the tumor microenvironment. Semin Immunopathol. 2018; 40: 545–554.
- 260Nagata S. Apoptosis and clearance of apoptotic cells. Annu Rev Immunol. 2018; 36: 489–517.
- 261Xi G, Gao J, Wan B, et al. GSDMD is required for effector CD8(+) T cell responses to lung cancer cells. Int Immunopharmacol. 2019; 74:105713.
- 262Hage C, Hoves S, Strauss L, et al. Sorafenib induces pyroptosis in macrophages and triggers natural killer cell-mediated cytotoxicity against hepatocellular carcinoma. Hepatology. 2019; 70: 1280–1297.
- 263Wang Q, Ren M, Feng F, Chen K, Ju X. Treatment of colon cancer with liver X receptor agonists induces immunogenic cell death. Mol Carcinog. 2018; 57: 903–910.
- 264Lu H, Zhang S, Wu J, et al. Molecular targeted therapies elicit concurrent apoptotic and GSDME-dependent pyroptotic tumor cell death. Clin Cancer Res. 2018; 24: 6066–6077.
- 265Gao Y, Zhang H, Zhou N, et al. Methotrexate-loaded tumour-cell-derived microvesicles can relieve biliary obstruction in patients with extrahepatic cholangiocarcinoma. Nat Biomed Eng. 2020; 4: 743–753.
- 266Fan JX, Deng RH, Wang H, et al. Epigenetics-based tumor cells pyroptosis for enhancing the immunological effect of chemotherapeutic nanocarriers. Nano Lett. 2019; 19: 8049–8058.
- 267Lu C, Guo C, Chen H, et al. A novel chimeric PD1-NKG2D-41BB receptor enhances antitumor activity of NK92 cells against human lung cancer H1299 cells by triggering pyroptosis. Mol Immunol. 2020; 122: 200–206.
- 268Ostrand-Rosenberg S, Beury DW, Parker KH, Horn LA. Survival of the fittest: how myeloid-derived suppressor cells survive in the inhospitable tumor microenvironment. Cancer Immunol Immunother. 2020; 69: 215–221.
- 269Jin S, Yang Z, Hao X, Tang W, Ma W, Zong H. Roles of HMGB1 in regulating myeloid-derived suppressor cells in the tumor microenvironment. Biomark Res. 2020; 8: 21.
- 270Parker KH, Horn LA, Ostrand-Rosenberg S. High-mobility group box protein 1 promotes the survival of myeloid-derived suppressor cells by inducing autophagy. J Leukoc Biol. 2016; 100: 463–470.
- 271Parker KH, Sinha P, Horn LA, et al. HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res. 2014; 74: 5723–5733.
- 272Park S, Cheon S, Cho D. The dual effects of interleukin-18 in tumor progression. Cell Mol Immunol. 2007; 4: 329–335.
- 273Zheng M, Kanneganti TD. The regulation of the ZBP1-NLRP3 inflammasome and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis). Immunol Rev. 2020; 297: 26–38.
- 274Orning P, Lien E. Multiple roles of caspase-8 in cell death, inflammation, and innate immunity. J Leukoc Biol. 2021; 109: 121–141.
- 275Reina-Campos M, Moscat J, Diaz-Meco M. Metabolism shapes the tumor microenvironment. Curr Opin Cell Biol. 2017; 48: 47–53.
- 276Dey P, Kimmelman AC, DePinho RA. Metabolic codependencies in the tumor microenvironment. Cancer Discov. 2021; 11: 1067–1081.
- 277Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol. 1927; 8: 519–530.
- 278Scharping NE, Rivadeneira DB, Menk AV, et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat Immunol. 2021; 22: 205–215.
- 279Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011; 331: 1565–1570.
- 280Van Acker HH, Ma S, Scolaro T, Kaech SM, Mazzone M. How metabolism bridles cytotoxic CD8(+) T cells through epigenetic modifications. Trends Immunol. 2021; 42: 401–417.
- 281Poznanski SM, Singh K, Ritchie TM, et al. Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment. Cell Metab 2021;; 33: 1205–1220.e5..
- 282Assmann N, O'Brien KL, Donnelly RP, et al. Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat Immunol. 2017; 18: 1197–1206.
- 283Liu X, Hartman CL, Li L, et al. Reprogramming lipid metabolism prevents effector T cell senescence and enhances tumor immunotherapy. Sci Transl Med. 2021; 13:eaaz6314.
- 284Ringel AE, Drijvers JM, Baker GJ, et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell. 2020; 183: 1848–1866.e1826.
- 285Iyengar NM, Gucalp A, Dannenberg AJ, Hudis CA. Obesity and cancer mechanisms: tumor microenvironment and inflammation. J Clin Oncol. 2016; 34: 4270–4276.
- 286Zitvogel L, Pietrocola F, Kroemer G. Nutrition, inflammation and cancer. Nat Immunol. 2017; 18: 843–850.
- 287Masters SL, Dunne A, Subramanian SL, et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nat Immunol. 2010; 11: 897–904.
- 288Rasheed A, Rayner KJ. Macrophage responses to environmental stimuli during homeostasis and disease. Endocr Rev. 2021. https://doi.org/10.1210/endrev/bnab004.
- 289Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018; 175: 998–1013.e1020.
- 290Madden MZ, Rathmell JC. The complex integration of T-cell metabolism and immunotherapy. Cancer Discov. https://doi.org/10.1158/2159-8290.CD-20-0569.
- 291Steven A, Fisher SA, Robinson BW. Immunotherapy for lung cancer. Respirology. 2016; 21: 821–833.
- 292Iams WT, Porter J, Horn L. Immunotherapeutic approaches for small-cell lung cancer. Nat Rev Clin Oncol. 2020; 17: 300–312.
- 293Jiang P, Gu S, Pan D, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018; 24: 1550–1558.
- 294Liu Y, Fang Y, Chen X, et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 2020; 5:eaax7969.
- 295Rotz SJ, Leino D, Szabo S, Mangino JL, Turpin BK, Pressey JG. Severe cytokine release syndrome in a patient receiving PD-1-directed therapy. Pediatr Blood Cancer. 2017; 64. https://doi.org/10.1002/pbc.26642..
- 296Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020; 70: 86–104.
- 297Hu J, Dong Y, Ding L, et al. Local delivery of arsenic trioxide nanoparticles for hepatocellular carcinoma treatment. Signal Transduct Target Ther. 2019; 4: 28.
- 298Serna N, Álamo P, Ramesh P, et al. Nanostructured toxins for the selective destruction of drug-resistant human CXCR4(+) colorectal cancer stem cells. J Control Release. 2020; 320: 96–104.