User:HY1998/sandbox

From Wikipedia, the free encyclopedia

Pyroptosis is a highly inflammatory form of programmed cell death that occurs most frequently upon infection with intracellular pathogens and is likely to form part of the antimicrobial response. This process promotes the rapid clearance of various bacterial, viral, fungal and protozoan infections by removing intracellular replication niches and enhancing the host's defensive responses. Pyroptosis can take place in immune cells and is also reported to occur in keratinocytes and some epithelial cells[1].

The process is initiated by formation of a large supramolecular complex termed the inflammasome (also known as a pyroptosome) upon intracellular danger signals. Inflammasome activates a different set of caspases compared to apoptosis, for example caspase-1/4/5 in human, and caspase-11 in mice.[2] These caspases contribute to the maturation and activation of several proinflammatory cytokines and pore forming protein gasdermins. Formation of pores causes cell membrane rupture and release of cytokines and various damage-associated molecular pattern (DAMP) molecules such as ATP and DNA out of the cell. These molecules recruit more immune cells and further perpetuate the inflammatory cascade in the tissue[3][4].

However, in pathogenic chronic diseases, the inflammatory response does not eradicate the primary stimulus, a chronic form of inflammation ensues that ultimately contributes to tissue damage. Pyroptosis is associated with diseases including cancer, neurodegeneration and cardiovascular diseases. Some examples of pyroptosis include Salmonella-infected macrophages and abortively HIV-infected T helper cells.[5] and abortively HIV-infected T helper cells.[6][7]

Discovery[edit]

This type of inherently proinflammatory programmed cell death was named 'pyroptosis' in 2001 by Dr. Brad T. Cookson, an associate professor of microbiology and laboratory medicine of University of Washington.[8] The Greek "pyro" refers to fire and "ptosis" means falling. The compound "pyroptosis" can be understood as "fiery falling", which describes the bursting of pro-inflammatory chemical signals from the dying cell. Pyroptosis has a distinct morphology and mechanism compared to other forms of cell death.[9] It was suggested that microbial infection was the main evolutionary pressure for this pathway.[10] In 2013, caspase-11 dependent noncanonical pathway was discovered, suggesting lipopolysaccharide (LPS) can trigger pyroptosis and subsequent inflammatory responses independent of Toll-like receptor 4 (TLR4)[11]. In 2015, gasdermin D was identified as the effector of pyroptosis that permeabilize the cell membrane[2][12].

Morphological Characteristics[edit]

Pyroptosis, as a form of programmed cell death, has many morphological differences compared to apoptosis. Both pyroptosis and apoptosis undergoes chromatin condensation, but during apoptosis, nucleus breaks into multiple chromatin bodies while in pyroptosis the nucleus remains intact[13]. The cell membrane remains intact during apoptosis. In a cell that undergoes pyroptosis, gasdermin pores are formed on the plasma membrane, resulting in water influx and cell lysis[1][14].

In terms of mechanism, pyroptosis is activated by inflammatory caspases including caspase-1/4/5 in human and caspase-11 in mice. Pro-apoptotic caspases, including caspase-6/ 7/8/9 are not required for pyroptosis. Caspase-3 activation notably, can take place in both apoptosis and pyroptosis[1][14].

Although both pyroptosis and necroptosis are


triggered by membrane pore formation, pyroptosis is more controlled. Cell that undergoes pyroptosis undergoes membrane blebbing and produces protrusions termed pyroptotic bodies, which is different from necroptosis[15]. Also, necroptosis works in a caspase independent fashion. It is proposed that both pyroptosis and necroptosis may act as defence systems against pathogens when apoptotic pathways are blocked.


Summary of the different morphologies, mechanisms, and outcomes of the 3 forms of cell death (apoptosis, pyroptosis, and necrosis)[9][16][14]
Characteristics Apoptosis Pyroptosis Necroptosis
Morphology Cell lysis NO YES YES
Cell swelling NO YES YES
Pore formation NO YES YES
Membrane blebbing YES YES NO
DNA fragmentation YES YES YES
Nucleus intact NO YES NO
Mechanism Caspase-1 activation NO YES NO
Caspase-3 activation YES YES NO
GSDMD activation NO YES NO
Outcome Inflammation NO YES YES
Programmed cell death YES YES YES

Mechanism[edit]

Innate immune system, by using germ-line encoded pattern recognition receptors (PRRs), can recognize a wide range of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) upon microbe infection. Classic examples of PRRs include Toll-like receptors (TLRs) and NOD-like receptors (NLRs)[17]. Recognition of PAMPs and DAMPs trigger the formation of multi-protein complex inflammasomes which then activates caspases to initiate pyroptosis. The inflammasome pathway can be canonical or noncanonical with the former using caspase-1 activating inflammasomes and the latter using other caspases[18].

The Canonical inflammasome pathway[edit]

In the canonical inflammasome pathway, PAMPs and DAMPs are recognised by certain endogenous PRRs. For example, NLR proteins NLRC4 can recognise flagellin and type III secretion system components[19]. NLRP3 is activated by cellular events induced by different PAMPs and DAMPs stimuli[20]. Some non-NLR proteins like absent in melanoma 2 (AIM2) and pyrin can also be activated and form inflammasomes[18]. Also, non-inflammasome forming PRRs like TLRs, NOD1 and NOD2 are also playing important roles in pyroptosis. These receptors upregulates expression of inflammatory cytokines such as IFN α/β, tumour necrosis factor (TNF), IL-6 and IL-12 through NF-κB and MAPK signaling pathways. In addition, pro-IL-1β and pro-IL-18 will be released to be processed by cysteine-mediated caspase-1[21][22].

Description of the image for the visually impaired
The formation of NLRP3 inflammasome

Canonical inflammasomes mostly contain 3 components, a sensor protein (PRRs), an adaptor (ASC), and an effector (caspase-1)[18]. Generally, inflammasome forming NLR proteins share a similar structure, several leucine-rich repeat (LRR) domains, a central nucleotide-binding and oligomerization domain (NBD) and an N-terminal pyrin domain (PYD). NLRP3, for example, recruits ASC adaptor protein via PYD-PYD interaction. Both pro-caspase-1 and ASC contains a caspase activation and recruitment domain (CARD) and this homotypic CARD-CARD interactions enables autocatalytic cleavage and reassembly of procaspase-1 to form active caspase-1[23]. Alternatively, NLRC4 can directly recruit pro-caspase-1 as it has a CARD domain instead of a pyrin domain[24].

Activated caspase-1 is responsible for cleavage of pro-IL-1β and pro-IL-18. These cytokines, once processed, will be in their biologically active form ready to be released from the host cells. In addition, caspase-1 also cleaves the cytosolic gasdermin D (GSDMD). GSDMD can be cleaved to produce an N-terminal domain (GSDMD-N) and a C-terminal domain (GSDMD-C). GSDMD-N can oligomerize and form transmembrane pores that have an inner diameter of 10-14nm[25]. The pores allow secretion of IL-1β and IL-18 and various cytosolic content to extracellular space. These pores also disrupts the cellular ionic gradient. The resulting increase in osmotic pressure causes an influx of water followed by cell swelling and bursting. Notably, GSDMD-N is autoinhibited by GSDMD C-terminal domain before cleavage to prevent cell lysis in normal conditions[26].Also, GSDMD-N can only insert itself into the inner membrane with specific lipid compositions[27], which limits its damage to neighbour cells.

The noncanonical inflammasome pathway[edit]

The noncanonical inflammasome pathway is initiated by binding of lipopolysaccharide (LPS) of gram-negative bacteria directly onto caspase-4/5 in human and caspase-11 in murines. Binding of LPS onto these caspases promotes their oligomerization and activation[28]. These caspases can cleave GSDMD to release GSDMD-N and triggers pyroptosis. In addition, influx of potassium ion upon membrane permeabilization triggers activation of NLRP3, which then leads to the formation of NLRP3 inflammasome and activation of caspase-1[18]. These processes facilitate the cleavage of GSDMD and promotes the maturation and release of proinflammatory cytokines.

Caspase-3 dependent pyroptosis pathway[edit]

An alternative pathway that links apoptosis and pyroptosis has been recently proposed. Caspase-3, an executioner caspase in apoptosis, can cleave gasdermin E (GSDME) to produce a N-terminal fragment and a C-terminal fragment in a way similar to GSDMD cleavage[2]. When apoptotic cell is not scavenged by macrophages, GSDME expression is then upregulated by p53. GSDME is then activated by caspase-3 to form pores on cell membrane. It is also found that GSDME can permeabilise mitochondrial membrane to release cytochrome c, which further activates caspase-3 and accelerates GSDME cleavage[29]. This positive feedback loop ensures programmed cell death to be carried forward.

Clinical relevance[edit]

Pyroptosis acts as a defence mechanism against infection by inducing pathological inflammation. The formation of inflammasomes and the activity of caspase-1 determine the balance between pathogen resolution and disease.

In a healthy cell, caspase-1 activation helps to fight infection caused by Salmonella and Shigella via introducing cell death to restrict pathogen growth.[16] When the ‘danger' signal is sensed, the quiescent cells will be activated to undergo pyroptosis and produce inflammatory cytokines IL-1β and IL-18. IL-18 will stimulate IFNγ production and initiates the development of TH1 responses. (TH1 responses tend to release cytokines that direct an immediate removal of the pathogen).[30] The cell activation results in an increase in cytokine levels, which will augment the consequences of inflammation and this, in turn, contributes to the development of the adaptive response as infection progresses. The ultimate resolution will clear pathogens.

In contrast, persistent inflammation will produce excessive immune cells which will be detrimental. If the amplification cycles persist, metabolic disorder, autoinflammatory diseases and liver injury associated with chronic inflammation will take place.[31]

Cancer[edit]

Pyroptosis, as an inflammation associated programmed cell death, has huge implications in various cancer types. Principally, pyroptosis can kill cancer cells and inhibit tumour develpment in the presence of endogenous DAMPs. In some cases, GSDMD can be used as a prognostic marker for cancers. However, prolonged production of inflammatory bodies may facilitate the formation of microenvironment that favours tumour growth[32]. Understanding mechanisms of pyroptosis and identification of pyroptosis associated molecules can be useful in treating different cancers.

In gastric cancer cells, presence of GSDMD can inhibit cyclin A2/CDK2 complexes, leading to cell cycle arrest and thus inhibits tumour development. Also, cellular concentration of GSDME increases when gastric cancer cells are treated with certain chemotherapy drugs. GSDME then activates caspase-3 and triggers pyroptotic cell death[14].

Cervical cancer can be caused by human papillomavirus (HPV) infection. AIM2 protein can recognise viral DNA in cytoplasm and form AIM2 inflammasome, which then triggers by caspase-1 dependent canonical pyroptosis pathway. HPV infection causes the upregulation of sirtuin 1 protein, which disrupt transcription factor for AIM2, RelB. Knockdown of sirtuin 1 upregulates AIM2 expression and triggers pyroptosis[33].

Metabolic disorder[edit]

The level of expression of NLRP3 inflammasome and caspase-1 has direct relation with the severity of several metabolic syndromes, such as obesity and type II diabetic mellitus (T2DM). This is because the subsequent production level of IL-1β and IL-18, cytokines that impairs the secretion of insulin, is affected by the activity of caspase-1. Glucose uptake level is then diminished, and the condition is known as insulin resistance.[34] The condition is further accelerated by the IL-1β induced destruction of pancreatic β cell.[35]

Cryopyrinopathies[edit]

A mutation in the gene coding of inflammasome leads to a group of autoinflammatory disease called cryopyrinopathies. This group includes Muckle–Wells syndrome, cold autoinflammatory syndrome, and chronic infantile neurologic cutaneous and articular syndrome, all showing symptoms of sudden fevers and localized inflammation.[36] The mutated gene in this case is the NLRP3, impeding the activation of inflammasome, resulting in an excessive production IL-1β. This effect is known as "gain-of-function".[37]

HIV and AIDS[edit]

Recent studies demonstrate that caspase-1-mediated pyroptosis drives CD4 T-cell depletion and inflammation by HIV,[38][39] two signature events that propel HIV disease progression to AIDS. Although pyroptosis contributes to the host's ability to rapidly limit and clear infection by removing intracellular replication niches and enhancing defensive responses through the release of proinflammatory cytokines and endogenous danger signals, in pathogenic inflammation, such as that elicited by HIV-1, this beneficial response does not eradicate the primary stimulus. In fact, it appears to create a pathogenic vicious cycle in which dying CD4 T cells release inflammatory signals that attract more cells into the infected lymphoid tissues to die and to produce chronic inflammation and tissue injury. It may be possible to break this pathogenic cycle with safe and effective caspase-1 inhibitors. These agents could form a new and exciting ‘anti-AIDS' therapy for HIV-infected subjects in which the treatment targets the host instead of the virus. Of note, Caspase-1 deficient mice develop normally,[40][41] arguing that inhibition of this protein would produce beneficial rather than harmful therapeutic effects in HIV patients.

References[edit]

  1. ^ a b c Jorgensen, Ine; Miao, Edward A (May 2015). "Pyroptotic cell death defends against intracellular pathogens". Immunological reviews. 265 (1): 130–42. doi:10.1111/imr.12287. PMID 25879289.
  2. ^ a b c Gong, Weihua; Shi, Ying; Ren, Jingjing (3 December 2019). "Research progresses of molecular mechanism of pyroptosis and its related diseases". Immunobiology: 151884. doi:10.1016/j.imbio.2019.11.019. PMID 31822435.
  3. ^ Baroja-Mazo A, Martín-Sánchez F, Gomez AI, Martínez CM, Amores-Iniesta J, Compan V, Barberà-Cremades M, Yagüe J, Ruiz-Ortiz E, Antón J, Buján S, Couillin I, Brough D, Arostegui JI, Pelegrín P (2014). "The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response". Nat Immunol. 15 (8): 738–48. doi:10.1038/ni.2919. PMID 24952504.
  4. ^ Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmüller W, Latz E (2014). "The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation". Nat Immunol. 15 (8): 727–37. doi:10.1038/ni.2913. PMC 4116676. PMID 24952505.
  5. ^ Fink SL, Cookson BT (November 2006). "Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages". Cell. Microbiol. 8 (11): 1812–25. doi:10.1111/j.1462-5822.2006.00751.x. PMID 16824040.
  6. ^ Doitsh, Gilad; Galloway, Nicole L. K.; Geng, Xin; Yang, Zhiyuan; Monroe, Kathryn M.; Zepeda, Orlando; Hunt, Peter W.; Hatano, Hiroyu; Sowinski, Stefanie; Muñoz-Arias, Isa; Greene, Warner C. (2014). "Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection". Nature. 505 (7484): 509–514. Bibcode:2014Natur.505..509D. doi:10.1038/nature12940. PMC 4047036. PMID 24356306.
  7. ^ Doitsh G, Greene WC (2016). "Dissecting How CD4 T Cells Are Lost During HIV Infection". Cell Host Microbe. 19 (3): 280–91. doi:10.1016/j.chom.2016.02.012. PMC 4835240. PMID 26962940.
  8. ^ Cookson, B.T& Brennan, M.A.; Brennan (2001). "Pro-inflammatory programmed cell death". Trends in Microbiology. 9 (3): 113–114. doi:10.1016/S0966-842X(00)01936-3.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  9. ^ a b L.Duprez; Berghe, Tom Vanden; Vandenabeele, Peter (2009). "Major cell death pathways at a glance". Microbes and Infection. 11 (13): 1050–62. doi:10.1016/j.micinf.2009.08.013. PMID 19733681. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)
  10. ^ M. Dagenais; A. Skeldon; M. Saleh (2012). "The inflammasome: in memory of Dr. Jurg Tschopp". Cell Death and Differentiation. 19 (1): 5–12. doi:10.1038/cdd.2011.159. PMC 3252823. PMID 22075986.
  11. ^ Shi, Jianjin; Zhao, Yue; Wang, Yupeng; Gao, Wenqing; Ding, Jingjin; Li, Peng; Hu, Liyan; Shao, Feng (6 August 2014). "Inflammatory caspases are innate immune receptors for intracellular LPS". Nature. 514 (7521): 187–192. doi:10.1038/nature13683. PMID 25119034.
  12. ^ Shi, Jianjin; Zhao, Yue; Wang, Kun; Shi, Xuyan; Wang, Yue; Huang, Huanwei; Zhuang, Yinghua; Cai, Tao; Wang, Fengchao; Shao, Feng (16 September 2015). "Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death". Nature. 526 (7575): 660–665. doi:10.1038/nature15514.
  13. ^ Brennan, Molly A.; Cookson, Brad T. (October 2000). "Salmonella induces macrophage death by caspase-1-dependent necrosis". Molecular Microbiology. 38 (1): 31–40. doi:10.1046/j.1365-2958.2000.02103.x.
  14. ^ a b c d Fang, Yuan; Tian, Shengwang; Pan, Yutian; Li, Wei; Wang, Qiming; Tang, Yu; Yu, Tao; Wu, Xi; Shi, Yongkang; Ma, Pei; Shu, Yongqian (January 2020). "Pyroptosis: A new frontier in cancer". Biomedicine & Pharmacotherapy. 121: 109595. doi:10.1016/j.biopha.2019.109595.
  15. ^ Chen, Xin; He, Wan-ting; Hu, Lichen; Li, Jingxian; Fang, Yuan; Wang, Xin; Xu, Xiaozheng; Wang, Zhuo; Huang, Kai; Han, Jiahuai (30 August 2016). "Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis". Cell Research. 26 (9): 1007–1020. doi:10.1038/cr.2016.100.
  16. ^ a b Susan L. Fink, Brad T. Cookson; Cookson (2006). "Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages". Cellular Microbiology. 8 (11): 1812–1825. doi:10.1111/j.1462-5822.2006.00751.x. PMID 16824040.
  17. ^ Karina R. Bortoluci & Ruslan Medzhitov; Medzhitov (2010). "Control of infection by pyroptosis and autophagy: role of TLR and NLR Cell". Biomedical & Life Sciences. 67 (10): 1643–1651. doi:10.1007/s00018-010-0335-5. PMID 20229126.
  18. ^ a b c d Platnich, Jaye M.; Muruve, Daniel A. (July 2019). "NOD-like receptors and inflammasomes: A review of their canonical and non-canonical signaling pathways". Archives of Biochemistry and Biophysics. 670: 4–14. doi:10.1016/j.abb.2019.02.008.
  19. ^ Zhao, Yue; Yang, Jieling; Shi, Jianjin; Gong, Yi-Nan; Lu, Qiuhe; Xu, Hao; Liu, Liping; Shao, Feng (14 September 2011). "The NLRC4 inflammasome receptors for bacterial flagellin and ype III secretion apparatus". Nature. 477 (7366): 596–600. doi:10.1038/nature10510.
  20. ^ Kelley, Nathan; Jeltema, Devon; Duan, Yanhui; He, Yuan (6 July 2019). "The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation". International Journal of Molecular Sciences. 20 (13): 3328. doi:10.3390/ijms20133328.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  21. ^ T Kawai1 and S Akira1 (2006). "TLR signaling". Cell Death and Differentiation. 13 (5): 816–825. doi:10.1038/sj.cdd.4401850. PMID 16410796.{{cite journal}}: CS1 maint: numeric names: authors list (link)
  22. ^ Mukherjee, Tapas; Hovingh, Elise Sofie; Foerster, Elisabeth G.; Abdel-Nour, Mena; Philpott, Dana J.; Girardin, Stephen E. (July 2019). "NOD1 and NOD2 in inflammation, immunity and disease". Archives of Biochemistry and Biophysics. 670: 69–81. doi:10.1016/j.abb.2018.12.022.
  23. ^ Broz, Petr; Dixit, Vishva M. (13 June 2016). "Inflammasomes: mechanism of assembly, regulation and signalling". Nature Reviews Immunology. 16 (7): 407–420. doi:10.1038/nri.2016.58.
  24. ^ Duncan, Joseph A.; Canna, Scott W. (January 2018). "The NLRC4 Inflammasome". Immunological Reviews. 281 (1): 115–123. doi:10.1111/imr.12607.
  25. ^ Ding, Jingjin; Wang, Kun; Liu, Wang; She, Yang; Sun, Qi; Shi, Jianjin; Sun, Hanzi; Wang, Da-Cheng; Shao, Feng (8 June 2016). "Pore-forming activity and structural autoinhibition of the gasdermin family". Nature. 535 (7610): 111–116. doi:10.1038/nature18590.
  26. ^ Liu, Zhonghua; Wang, Chuanping; Rathkey, Joseph K.; Yang, Jie; Dubyak, George R.; Abbott, Derek W.; Xiao, Tsan Sam (May 2018). "Structures of the Gasdermin D C-Terminal Domains Reveal Mechanisms of Autoinhibition". Structure. 26 (5): 778–784.e3. doi:10.1016/j.str.2018.03.002.
  27. ^ Qiu, Shiqiao; Liu, Jing; Xing, Feiyue (3 March 2017). "'Hints' in the killer protein gasdermin D: unveiling the secrets of gasdermins driving cell death". Cell Death & Differentiation. 24 (4): 588–596. doi:10.1038/cdd.2017.24.
  28. ^ Shi, Jianjin; Zhao, Yue; Wang, Yupeng; Gao, Wenqing; Ding, Jingjin; Li, Peng; Hu, Liyan; Shao, Feng (6 August 2014). "Inflammatory caspases are innate immune receptors for intracellular LPS". Nature. 514 (7521): 187–192. doi:10.1038/nature13683.
  29. ^ Rogers, Corey; Erkes, Dan A.; Nardone, Alexandria; Aplin, Andrew E.; Fernandes-Alnemri, Teresa; Alnemri, Emad S. (11 April 2019). "Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation". Nature Communications. 10 (1). doi:10.1038/s41467-019-09397-2.
  30. ^ Nakanishi, K., Yoshimoto, T., Tsutsui, H. & Okamura, H.; Wen; Ting (2011). "The Inflammasome NLRs in Immunity, Inflammation, and Associated Diseases". Annual Review of Immunology. 29 (1): 707–735. doi:10.1146/annurev-immunol-031210-101405. PMC 4067317. PMID 21219188.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  31. ^ B.K. Davis, HaitaoWen & J.P.-Y. Ting; Wen; Ting (2011). "The Inflammasome NLRs in Immunity, Inflammation, and Associated Diseases". Annual Review of Immunology. 29 (1): 707–735. doi:10.1146/annurev-immunol-031210-101405. PMC 4067317. PMID 21219188.
  32. ^ Xia, Xiaojing; Wang, Xin; Cheng, Zhe; Qin, Wanhai; Lei, Liancheng; Jiang, Jinqing; Hu, Jianhe (9 September 2019). "The role of pyroptosis in cancer: pro-cancer or pro-"host"?". Cell Death & Disease. 10 (9). doi:10.1038/s41419-019-1883-8.
  33. ^ So, Daeho; Shin, Hyun-Woo; Kim, Jiyoung; Lee, Mingyu; Myeong, Jongyun; Chun, Yang-Sook; Park, Jong-Wan (29 May 2018). "Cervical cancer is addicted to SIRT1 disarming the AIM2 antiviral defense". Oncogene. 37 (38): 5191–5204. doi:10.1038/s41388-018-0339-4.
  34. ^ Vandanmagsar, B. (2011). "The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance". Nature Medicine. 17 (2): 179–188. doi:10.1038/nm.2279. PMC 3076025. PMID 21217695. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)
  35. ^ T.Strowig, J.Henao-Mejia, E.Elinav & R.Flavell; Henao-Mejia; Elinav; Flavell (2012). "Inflammasomes in health and disease". Nature. 481 (7381): 278–286. Bibcode:2012Natur.481..278S. doi:10.1038/nature10759. PMID 22258606.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  36. ^ B. Neven; A. Prieur; P. Quartier dit Maire (2008). "Cryopyrinopathies: update on pathogenesis and treatment". Nature Clinical Practice Rheumatology. 4 (9): 481–489. doi:10.1038/ncprheum0874. PMID 18665151.
  37. ^ L.D Church; G.P Cook; M.F McDermott (2008). "Primer: inflammasomes and interleukin 1β in inflammatory disorders". Nature Clinical Practice Rheumatology. 4 (1): 34–42. doi:10.1038/ncprheum0681. PMID 18172447.
  38. ^ Doitsh, Gilad; Galloway, Nicole L. K.; Geng, Xin; Yang, Zhiyuan; Monroe, Kathryn M.; Zepeda, Orlando; Hunt, Peter W.; Hatano, Hiroyu; Sowinski, Stefanie; Muñoz-Arias, Isa; Greene, Warner C. (2014). "Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection". Nature. 505 (7484): 509–14. Bibcode:2014Natur.505..509D. doi:10.1038/nature12940. PMC 4047036. PMID 24356306.
  39. ^ Monroe, K. M.; Yang, Z; Johnson, J. R.; Geng, X; Doitsh, G; Krogan, N. J.; Greene, W. C. (2014). "IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV". Science. 343 (6169): 428–32. Bibcode:2014Sci...343..428M. doi:10.1126/science.1243640. PMC 3976200. PMID 24356113.
  40. ^ Kuida, K; Lippke, J. A.; Ku, G; Harding, M. W.; Livingston, D. J.; Su, M. S.; Flavell, R. A. (1995). "Altered cytokine export and apoptosis in mice deficient in interleukin-1 beta converting enzyme". Science. 267 (5206): 2000–3. Bibcode:1995Sci...267.2000K. doi:10.1126/science.7535475. PMID 7535475.
  41. ^ Li, P; Allen, H; Banerjee, S; Franklin, S; Herzog, L; Johnston, C; McDowell, J; Paskind, M; Rodman, L; Salfeld, J (1995). "Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock". Cell. 80 (3): 401–11. doi:10.1016/0092-8674(95)90490-5. PMID 7859282.

Category:Programmed cell death