Open Access

The NLRP3 inflammasome in viral infection (Review)

  • Authors:
    • Qiaoli Zheng
    • Chunting Hua
    • Qichang Liang
    • Hao Cheng
  • View Affiliations

  • Published online on: July 6, 2023     https://doi.org/10.3892/mmr.2023.13047
  • Article Number: 160
  • Copyright: © Zheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The interplay between pathogen and host determines the immune response during viral infection. The Nod‑like receptor (NLR) protein 3 inflammasome is a multiprotein complex that induces the activation of inflammatory caspases and the release of IL‑1β, which play an important role in the innate immune responses. In the present review, the mechanisms of the NLR family pyrin domain containing 3 inflammasome activation and its dysregulation in viral infection were addressed.

Introduction

Inflammasomes are a group of multiprotein complexes that recognize both extracellular and cytoplasmic intracellular pathogens and danger signals. The assembly of the specific inflammasomes is induced by various pattern-recognition receptors (PRRs) in response to pathogen associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), and results in the induction of pro-inflammatory cytokines, such as IL-1β and IL-18 (13). In addition, inflammasomes have been proposed to regulate other key events in inflammation and tissue repair, such as pyroptosis, a highly inflammatory form of programmed cell death. Therefore, the inflammasomes play an important role in a variety of human pathophysiological processes, including antimicrobial response and development of metabolic syndromes, cancer, autoimmune and neurodegenerative diseases (4).

To date, the following five types of PRRs have been identified: Toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors, C-type lectin receptors (CLRs), DNA sensors [DNA-dependent activator of IFN-regulatory factors and absent in melanoma 2], and the nucleotide-binding domain (NBD), leucine-rich repeat-containing (LRR) proteins (NLRs) (1,2). Several studies have demonstrated that the NLR family includes NLR proteins (NLRP) 1, 2, 3, 6, 12, the NLR family caspase activation and recruitment domain (CARD), and the nucleotide-binding oligomerization domain-containing protein 2 (59). Among them, the NLRP3 inflammasome can be activated by a wide variety of stimuli, including DAMPs, PAMPs and bacterial toxins. In the present review, the biological process of the NLRP3 inflammasome assembly, its activation, and the recent developments of its involvement in viral infection are reviewed. The findings highlight the research challenges and future directions on this scientific field.

The NLRP3 inflammasome

NLR structure

NLRs consist of three separate domains, including an N-terminal pyrin domain (PYD), a central NBD, and a C-terminal LRR (Fig. 1). The N terminal domain is a caspase recruitment domain, a CARD, or a baculovirus inhibitory repeat domain, which has been applied as a structural subclassification of the NLR family (1,10). NLRP3 (NLR family, pyrin domain containing 3), which is also called cryopyrin, NALP3, CIAS1, CLR1.1, and PYPAF1 contains a PYD, a central NACHT (nucleotide-binding domain or NAIP, CIITA, HET-E and TP1) and a C-terminal LRR, which lacks CARD and cannot recruit pro-caspase-1 in the absence of the adaptor molecule caspase recruitment domain (ASC) (1). The NACHT domain is primarily responsible for dNTPase activity and oligomerization, while the PYD domain mediates downstream signaling of homotypic protein-protein interactions. LRR has been involved in ligand-sensing and autoregulation. All three domains are involved in the protein interaction networks (7).

NLRP3 complex

In addition to the inflammasome sensor molecules, ASCs are present that connect caspase 1 in the NLRP3 inflammasome complex. ASC contains two death-fold domains: One PYD and one CARD. The interaction between the upstream inflammasome sensor molecules triggers ASC assembly via the pyrin domain into a large protein complex (11,12). ASC brings procaspase 1 into close proximity with CARD, which initiates caspase 1 self-cleavage and activation (13,14).

Caspase 1

The cysteine protease caspase 1 is a key player of the inflammatory response. Procaspase-1 recruitment to the ASC speck enables its dimerization and autoactivation (1). The CARD domain is separated by a CARD domain linker from the C-terminal catalytic domain, containing large (p20) and small (p10) subunits (15). A cleavage product, p33 (CARD + p20), is generated by the interdomain linker between p20 and p10. It is confirmed that the active species of caspase 1 in macrophages is a transient tetramer composed of p33 and p10 subunits (p33/p10). The complex removes the CARD domain by self-cleavage, leading to the release of p20/p10 and the loss of enzymatic activity (15).

IL-1β and IL-18

Active caspase 1 further processes pro-IL-1β and pro-IL-18 into their mature forms. Serine proteinases including cathepsin G, elastase, and proteinase 3 can cleave pro-IL-1β except for caspase-1 (16). Furthermore, mast cell chymase is able to cleave pro-IL-18 (17). IL-1 family cytokines are key initiators and regulators of immune responses and inflammation. IL-1β and IL-18 are significant cytokines that regulate innate and adaptive immune responses (18). IL-18 plays a major role in the induction of the Th1 response, while IL-1β contributes to the T-helper 17 response (19). In various cases, the secretion of IL-1 cytokines is closely linked to cell death. Previous studies have suggested that cell lysis is the primary release mechanism for IL-1β and IL-18 (19,20). However, recently, a common secretory pathway in the absence of cell death and cell lysis was identified, which depended on membrane permeability (20). The secretion of IL-1β and IL-18 indicates the important role of the NLRP3 inflammasome in inflammatory and infectious diseases.

Except for the cytokine release, inflammasomes also trigger pyroptosis, which is induced by inflammatory caspases (murine caspase 1 and caspase 11 or human caspase 4 and caspase 5) (21). Pyroptosis is a necrotic form of infected cell death that is different from classical apoptosis or necrosis and is characterized by cell swelling, osmotic lysis and the subsequent release of intracellular content (21). Gasdermin D is required for the induction of pyroptosis and its N-terminal fragment indicates the intrinsic pyroptosis-inducing activity (22).

Activation and regulation of the NLRP3 inflammasome

NLRP3 is known to respond to a variety of stimuli, including PAMPs, DAMPs and bacterial toxins. These PAMPs include fungi, such as Candida albicans and Saccharomyces cerevisiae (23); bacteria, including Staphylococcus aureus (S. aureus), Listeria monocytogenes and Neisseria gonorrhoeae (2426); and viruses, such as Sendai, influenza, encephalomyocarditis viruses and adenovirus (7,2729). In addition, several host-derived molecules, which are indicative of cellular injury or stress can also activate the NLRP3 inflammasome, including ATP released from necrotic cells, hyaluronan, glucose, monosodium urate, myeloid-β, skin irritants, imidazoquinoline compounds, silica, asbestos and alum (Fig. 2) (30). In certain cases, the inflammasome is activated by individual microbial components, such as, the α-toxin of S. aureus (24,3133).

Canonical activation

Despite these findings, a universal activation mechanism for NLRP3 has not been reported. Extracellular ATP stimulates the P2X purinoceptor 7, triggering efflux of potassium and inducing the progressive recruitment of the membrane pore pannexin-1 (34). Under the second model, phagocytes engulf the inflammasomal activators, such as silica, monosodium urate, amyloid-β, asbestos, and alum, leading to lysosomal damage. The cytosolic release of lysosomal contents is sensed by the NLRP3 inflammasome (35,36). At last, nearly all NLRP3 agonists induce the generation of reactive oxygen species (ROS) (3739). ROS blockade by chemical scavengers inhibit NLRP3 inflammasome activation (23,40,41). In addition, mitochondrial dysfunction induces changes in intracellular calcium levels, and the release of oxidized mitochondrial DNA can lead to NLRP3 activation (42).

ROS

Inflammasome activation can be triggered by ROS; however, the exact mechanism remains unclear. A nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is involved in the process, since NLRP3 inflammasome is inhibited upon suppression of NADPH oxidase common p22 subunit (39). A recent study suggests that thioredoxin-interacting protein (TXNIP/VDUP1) is implicated in NLRP3 activation. Inflammasome activators induce the ROS-dependent TXNIP dissociation from thioredoxin. TXNIP further binds to NLRP3 and its deficiency impairs activation of the inflammasome and subsequent secretion of IL-1β (30).

Oxidized mitochondrial DNA (mtDNA)

The oxidized mtDNA from stressed mitochondria is also suggested to be the cytoplasmic activator of the NLRP3 inflammasome. A previous study conducted by Shimada et al (43) revealed that oxidized mtDNA could bind directly and activate the NLRP3 inflammasome, while macrophages lacking mtDNA severely reduced IL-1β production. In addition, ROS may also be of mitochondrial origin. However, whether ROS or oxidized mtDNA acts as a direct activator of the NLRP3 inflammasome or as its cofactor remains unknown.

Non-canonical activation

In complement with the canonical inflammasome pathway, the non-canonical pathway is mediated by murine caspase 11 and caspase 4 or caspase 5 in human cells in response to Gram-negative bacteria (4447). Lipopolysaccharide (LPS, a component of the Gram-negative bacterial cell wall) was shown to induce the activation of caspase 11 in mice and of caspase 4 or caspase 5 in human cells, which was previously attributed to the LPS receptor TLR4 (4547). However, recent studies have shown that host cells have developed TLR4-independent mechanisms to recognize the cytoplasmic LPS. It was found that TLR4(−/-) mice primed with the TLR3 agonist polyinosinic: Polycytidylic acid induced pro-caspase-11 expression, which could possibly explain this discrepancy (4547). The activation of caspases 4, 5 and 11 initiates pyroptosis similarly to caspase 1, which is not responsible for cleaving pro-IL-1β or pro-IL-18 (44,48). Upon activation, these caspases also promote the assembly of the NLRP3 inflammasome (44,49,50).

In the absence of microbial stimulation, exposure to TNF-α significantly promotes ATP or silica-mediated caspase 1 activation and IL-1 secretion in macrophages and dendritic cells (51). Signals provided by NF-κB activators are not sufficient for NLRP3 activation (52). Certain studies have shown that NIMA-related kinase 7 is an important component of the NLRP3 complex, which is required for NLRP3 activation, responding to both canonical and non-canonical stimuli (5355). The kinase spleen tyrosine kinase (SYK) is essential for NLRP3 inflammasome activation (23,41).

Positive regulation

Over the past decade, numerous studies have uncovered the role and regulation of inflammasomes during certain pathophysiological processes. The regulatory mechanisms include transcriptional, post-transcriptional regulation, post-translational modifications and regulatory proteins that target the receptors, ASC or the caspases. Furthermore, several other studies have highlighted the microbial evolution of inflammasome inhibitors.

The integration with PRRs or cytokine receptors highly influences the NLRP3 inflammasome activation and pro-IL-1β availability (51,52). In response to PRR stimulation, NLRP3 deubiquitylation, which is mediated by the K63-specific deubiquitinase BRCA1-BRCA2-containing complex subunit 3, also induces inflammasome signaling (56,57). The double-stranded RNA-dependent protein kinase R or eukaryotic translation initiation factor 2-α kinase 2 was shown to control NLRP3 inflammasome activity (58). Selective activation of the NLRP3 inflammasome was promoted by guanylate binding protein 5, in responses to pathogenic and soluble bacteria but not in response to crystalline inflammasome priming substances (59). These data suggest that small heterodimer partner is a key mediator in controlling NLRP3 inflammasome activation via mechanisms that interact with NLRP3 and maintain mitochondrial homeostasis (60).

The SYK and JNK enzymes phosphorylate the inflammasome adaptor ASC, which further contributes to the activation of caspase 1 (61). It has also been shown that SYK promotes NLRP3-dependent caspase 1 activation during infections with Candida albicans (23). The TGFβ-activated kinase 1 (TAK1 or MAP3K7)-JNK pathway was activated by ruptured lysosomes and promoted NLRP3 inflammasome activation, through the oligomerization of an adapter protein and apoptosis-associated speck-like protein including ASC (62).

Negative regulation

A number of proteins containing a CARD or a PYD have been proposed to inhibit inflammasome activity by blocking inflammasome component recruitment. Certain CARD-only proteins (COPs), such as human CARD16 (also known as COP or PSEUDO-ICE), CARD17 (also known as INCA), and CARD18 (also known as ICEBERG), are highly similar to the CARD of caspase 1 (6365). They are suggested to suppress inflammasome activation by sequestering caspase 1. In addition, they may also regulate other signaling pathways. For instance, CARD16 and CARD18 activate receptor-interacting serine/threonine protein kinase 2 (6365). Caspase 12 is an inhibitor of caspase 1 and its overexpression appears to abrogate caspase 1 activity (66).

PYD-only proteins (POPs), including pyrin, POP1 (PYDC1), POP2 (PYDC2) and POP3 (PYDC5), regulate inflammasome signaling at the molecular level of the PYD-PYD interaction (6770). It was shown that binding of POP1 to ASC inhibited ASC-dependent inflammasome assembly by preventing inflammasome nucleation, and consequently reducing IL-1β release. Although mice do not have the ortholog of POP1, its transgenic expression can protect them from PAMP-triggered inflammation. Moreover, POP1 was regulated by TLR and IL-1 receptor signaling; therefore, it was proposed that POP1 would provide a regulatory feedback loop and shut down excessive inflammatory responses (70). An additional member of POPs, POP2, binds to ASC and PAN1, and inhibits the formation of cryopyrin and inflammasomes (68). In addition, it also inhibits NF-κB activation (67). The other PYD-only protein, POP3 was identified to compete with ASC for recruitment of liver regeneration-associated proteins, further reducing inflammasome activation (69).

The kinase IκB kinase-α, is involved in the negative regulation of ASC by controlling its subcellular localization in resting macrophages; however, it is not clear whether this process is associated with ASC phosphorylation (71).

Viral infection and NLRP3 inflammasome

The NLRP3 inflammasome is critical in the host anti-viral immune function. Previous studies have shown that infection from both RNA and DNA viruses can induce activation of the NLRP3 inflammasome and promote the secretion of IL-1β and IL-18 (72), such as Rift Valley fever virus, encephalomyocarditis virus (EMCV), foot-and-mouth disease virus, Mayaro virus (MAYV), hepatitis B and C viruses, Zika virus (ZIKV), H7N9 influenza A virus, Dengue virus and varicella-zoster virus (7280). When viruses invade their hosts, the NLRP3 inflammasome can be activated by sensing viral components, including RNA, DNA and proteins. However, the mechanisms of the NLRP3 inflammasome activation during a viral infection are still debatable.

Viroporins are a group of low-molecular-weight proteins, which are important for innate immune responses, notably for the activation of the NLRP3 inflammasome, such as P7 viroporin of the hepatitis C virus, viroporin 2B of EMCV, and envelope (E) protein of severe acute respiratory syndrome coronavirus viroporin 3a (72,8183). During the infection, viroporins can increase the cell membrane permeability, facilitate viral invasion, and cause imbalance of ion concentration levels, such as Ca2+, which in turn produces ion fluxes and promotes the activation of NLRP3 (84). Angiotensin converting enzyme 2, which is expressed in injured type II alveolar epithelial cells may lead to NLRP3 activation during coronavirus disease (COVID)-19 infection (85).

A subsequent study revealed that certain infections from RNA viruses could cause potassium efflux, which is a trigger for the activation of the NLRP3 inflammasome (86). Infection by murine hepatitis viral strain-3 can induce the quick release of ROS, which may act as a trigger for NLRP3 inflammasome activation (87). ROS also plays an important role in human immunodeficiency virus (HIV)-1 infection. HIV-1 will promote ROS production and induce inflammasome activation (88). MAYV infection can induce ROS release and potassium efflux, which subsequently triggers the activation of the NLRP3 inflammasome. Human parainfluenza virus type 3 infection can induce NLRP3 inflammasome activation via TLR2 activation and potassium efflux (89). The M2 and PB1-F2 proteins of the influenza virus can activate the NLRP3 inflammasome by regulating intracellular ionic and mitochondrial ROS production. During this process, oxidized DNA plays an important role and influenza viruses can induce oxidized DNA release (90). Influenza viral infection will also induce lysosomal damage through the lysosomal pathway, further activating NLRP3 (29). Adenoviral infections release lysosomal cathepsin B into the cytoplasm, which can cause the activation of the NLRP3 inflammasome (91).

The RNase L system can sense double-stranded RNAs and generate RNA cleavage products to activate the NLRP3 inflammasome. DExD/H-box helicase and the mitochondria-associated molecule mitochondrial antiviral-signaling protein (MAVS) participate in this process (92). MAVS can recruit NLRP3 to the mitochondria and promote the secretion of IL-1β (93). DEAD/H-box RNA helicases (DDX), DDX33 and DDX19A act as RNA sensors involved in NLRP3 inflammasome activation (94). The RNA helicase RIG-I acts as a sensor to certain RNA viruses, such as vesicular stomatitis and influenza viruses, through the NF-κB pathway to activate the NLRP3 inflammasome (29). Certain RNA viruses activate NLRP3 via the RIP1-receptor-interacting serine/threonine-protein kinase 3 (RIP3)-DRP1 pathway. The RIP1-RIP3 complex is assembled, which induces mitochondrial damage and activates NLRP3 (95).

Viruses have also evolved multiple mechanisms to evade the host immune response. Certain viral proteins can suppress the NLRP3 inflammasome-associated immune response, such as the V protein of measles virus. Sendai virus can inhibit activation of the NLRP3 inflammasome and reduce secretion of IL-1β by interacting with NLRP3 (96,97). The influenza A viral NS1 protein suppresses ASC ubiquitination and speck formation via the RIG-I/type I IFN pathway to inhibit NLRP3 inflammasome activation and reduce the production of IL-1β (98,99). The PB1-F2 protein of avian influenza A suppresses NLRP3 inflammasome-dependent IL-1β secretion by targeting the MAVS-NLRP3 interaction (100). RNA I of adenovirus VA inhibits the activation of the NLRP3 inflammasome by suppressing ASC phosphorylation and oligomerization (101). The non-structural protein NS3 of ZIKV reduces NLRP3-mediated IL-1β secretion (102). An Epstein-Barr virus microRNA (miR) can inhibit the NLRP3 inflammasome by targeting the miR-223 site (103). Enterovirus 71 can cleave NLRP3 by activation of proteases 2A and 3C, which will inhibit NLRP3 and IL-1β secretion (104). Hepatitis B viral infection can inhibit NF-κB phosphorylation and ROS production. Hepatitis B e-antigen suppresses the activation of NLRP3 and IL-1β production (105). Herpes simplex virus 1 has evolved mechanisms to block the NLRP3 activity (106).

Conclusion

The NLRP3 inflammasome has been widely studied in innate immune responses. However, the exact mechanisms of NLRP3 inflammasome activation and regulation remain unclear, notably during the viral infection process. In the present review, the current knowledge of the NLRP3 assembly process and its regulatory mechanisms during viral infection were summarized. The findings indicate that viruses can evade the host immune response by inhibiting the NLRP3 cytokine storm. As a result, it is of value to elucidate the immunological balance and the underlying mechanism of this process. Certain studies have supported the hypothesis that targeting the NLRP3 pathway can be used in the development of therapeutic strategies for viral infection, notably for COVID-19. The development of antiviral therapies requires additional studies that will explore the molecular pathogenesis of NLRP3 inflammasome activation in response to viral infection.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Young Scientists Fund of the National Natural Science Foundation of China (grant no. 81801992), and the Medical and Health Science and Technology Project of Zhejiang (grant nos. 2018256428 and 2022514972).

Availability of data and materials

Not applicable.

Authors' contributions

QZ and HC conceived and designed the study. QZ, CH and QL collected data. QZ, CH and QL prepared the draft of the manuscript. HC reviewed and edited the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Schroder K and Tschopp J: The inflammasomes. Cell. 140:821–832. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Fusco R, Siracusa R, Genovese T, Cuzzocrea S and Di Paola R: Focus on the role of NLRP3 inflammasome in diseases. Int J Mol Sci. 21:42232020. View Article : Google Scholar : PubMed/NCBI

3 

Farag NS, Breitinger U, Breitinger HG and El Azizi MA: Viroporins and inflammasomes: A key to understand virus-induced inflammation. Int J Biochem Cell Biol. 122:1057382020. View Article : Google Scholar : PubMed/NCBI

4 

Kelley N, Jeltema D, Duan Y and He Y: The NLRP3 inflammasome: An overview of mechanisms of activation and regulation. Int J Mol Sci. 20:33282019. View Article : Google Scholar : PubMed/NCBI

5 

Barnett KC, Li S, Liang K and Ting JPY: A 360° view of the inflammasome: Mechanisms of activation, cell death, and diseases. Cell. 186:2288–2312. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Chou WC, Jha S, Linhoff MW and Ting JPY: The NLR gene family: from discovery to present day. Nat Rev Immunol. Mar 27–2023.(Epub ahead of print). View Article : Google Scholar

7 

Chai R, Li Y, Shui L, Ni L and Zhang A: The role of pyroptosis in inflammatory diseases. Front Cell Dev Biol. 11:11732352023. View Article : Google Scholar : PubMed/NCBI

8 

Zhan X, Li Q, Xu G, Xiao X and Bai Z: The mechanism of NLRP3 inflammasome activation and its pharmacological inhibitors. Front Immunol. 13:11099382023. View Article : Google Scholar : PubMed/NCBI

9 

Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN and Tschopp J: NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 20:319–325. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Song JY, Park YM and Choi SY: Type 2 human papillomavirus E7 attenuates E-cadherin expression in human keratinocytes. J Microbiol. 59:616–625. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Yu TG, Cha JS, Kim G, Sohn YK, Yoo Y, Kim U, Song JJ, Cho HS and Kim HS: Oligomeric states of ASC specks regulate inflammatory responses by inflammasome in the extracellular space. Cell Death Discov. 9:1422023. View Article : Google Scholar : PubMed/NCBI

12 

Proell M, Gerlic M, Mace PD, Reed JC and Riedl SJ: The CARD plays a critical role in ASC foci formation and inflammasome signalling. Biochem J. 449:613–621. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Guo Y, Gu D, Huang T, Li A, Zhou Y, Kang X, Meng C, Xiong D, Song L, Jiao X and Pan Z: Salmonella Enteritidis T1SS protein SiiD inhibits NLRP3 inflammasome activation via repressing the mtROS-ASC dependent pathway. PLoS Pathog. 19:e10113812023. View Article : Google Scholar : PubMed/NCBI

14 

de Souza JG, Starobinas N and Ibañez OCM: Unknown/enigmatic functions of extracellular ASC. Immunology. 163:377–388. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Ross C, Chan AH, von Pein JB, Maddugoda MP, Boucher D and Schroder K: Inflammatory caspases: Toward a unified model for caspase activation by inflammasomes. Annu Rev Immunol. 40:249–269. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Triantafilou K: Enigmatic inflammasomes. Immunology. 162:249–251. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Wang C, Yang T, Xiao J, Xu C, Alippe Y, Sun K, Kanneganti TD, Monahan JB, Abu-Amer Y, Lieberman J and Mbalaviele G: NLRP3 inflammasome activation triggers gasdermin D-independent inflammation. Sci Immunol. 6:eabj38592021. View Article : Google Scholar : PubMed/NCBI

18 

Zhen Y and Zhang H: NLRP3 inflammasome and inflammatory bowel disease. Front Immunol. 10:2762019. View Article : Google Scholar : PubMed/NCBI

19 

Voet S, Srinivasan S, Lamkanfi M and van Loo G: Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med. 11:e102482019. View Article : Google Scholar : PubMed/NCBI

20 

Tapia VS, Daniels MJD, Palazón-Riquelme P, Dewhurst M, Luheshi NM, Rivers-Auty J, Green J, Redondo-Castro E, Kaldis P, Lopez-Castejon G and Brough D: The three cytokines IL-1β, IL-18, and IL-1α share related but distinct secretory routes. J Biol Chem. 294:8325–8335. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Long J, Sun Y, Liu S, Yang S, Chen C, Zhang Z, Chu S, Yang Y, Pei G, Lin M, et al: Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov. 9:1552023. View Article : Google Scholar : PubMed/NCBI

22 

Devant P and Kagan JC: Molecular mechanisms of gasdermin D pore-forming activity. Nat Immunol. Jun 5–2023.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

23 

Li Y, Song W, Tong Y, Zhang X, Zhao J, Gao X, Yong J and Wang H: Isoliquiritin ameliorates depression by suppressing NLRP3-mediated pyroptosis via miRNA-27a/SYK/NF-κB axis. J Neuroinflammation. 18:12021. View Article : Google Scholar : PubMed/NCBI

24 

Mariathasan S, Weiss DS, Newton K, McBride J, O'Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM and Dixit VM: Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 440:228–232. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Guo Y, Li L, Xu T, Guo X, Wang C, Li Y, Yang Y, Yang D, Sun B, Zhao X, et al: HUWE1 mediates inflammasome activation and promotes host defense against bacterial infection. J Clin Invest. 130:6301–6316. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Zhang S, Zhang Y, Gan L, Wei F, Chai B, A Aljaafreh AAH, Liu X, Duan X, Jiang J, Wang X, et al: Progesterone suppresses Neisseria gonorrhoeae-induced inflammation through inhibition of NLRP3 inflammasome pathway in THP-1 cells and murine models. Front Microbiol. 12:5700932021. View Article : Google Scholar : PubMed/NCBI

27 

Muruve DA, Pétrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, Parks RJ and Tschopp J: The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 452:103–107. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Allen IC, Scull MA, Moore CB, Holl EK, McElvania-TeKippe E, Taxman DJ, Guthrie EH, Pickles RJ and Ting JP: The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 30:556–565. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Poeck H, Bscheider M, Gross O, Finger K, Roth S, Rebsamen M, Hannesschläger N, Schlee M, Rothenfusser S, Barchet W, et al: Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1 beta production. Nat Immunol. 11:63–69. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Zhou R, Tardivel A, Thorens B, Choi I and Tschopp J: Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 11:136–140. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Walev I, Reske K, Palmer M, Valeva A and Bhakdi S: Potassium-inhibited processing of IL-1 beta in human monocytes. EMBO J. 14:1607–1614. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Gurcel L, Abrami L, Girardin S, Tschopp J and van der Goot FG: Caspase-1 activation of lipid metabolic pathways in response to bacterial pore-forming toxins promotes cell survival. Cell. 126:1135–1145. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Liu R, Liu Y, Liu C, Gao A, Wang L, Tang H, Wu Q, Wang X, Tian D, Qi Z and Shen Y: NEK7-mediated activation of NLRP3 inflammasome is coordinated by potassium Efflux/Syk/JNK signaling during Staphylococcus aureus infection. Front Immunol. 12:7473702021. View Article : Google Scholar : PubMed/NCBI

34 

Pelegrin P: P2X7 receptor and the NLRP3 inflammasome: Partners in crime. Biochem Pharmacol. 187:1143852021. View Article : Google Scholar : PubMed/NCBI

35 

Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ and Golenbock DT: The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 9:857–865. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA and Latz E: Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 9:847–856. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Cruz CM, Rinna A, Forman HJ, Ventura ALM, Persechini PM and Ojcius DM: ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem. 282:2871–2879. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Cassel SL, Eisenbarth SC, Iyer SS, Sadler JJ, Colegio OR, Tephly LA, Carter AB, Rothman PB, Flavell RA and Sutterwala FS: The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci USA. 105:9035–9040. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT and Tschopp J: Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 320:674–677. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Koumangoye R: The role of Cl and K+ efflux in NLRP3 inflammasome and innate immune response activation. Am J Physiol Cell Physiol. 322:C645–C652. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Shio MT, Eisenbarth SC, Savaria M, Vinet AF, Bellemare MJ, Harder KW, Sutterwala FS, Bohle DS, Descoteaux A, Flavell RA and Olivier M: Malarial hemozoin activates the NLRP3 inflammasome through Lyn and Syk kinases. PLoS Pathog. 5:e10005592009. View Article : Google Scholar : PubMed/NCBI

42 

Latz E, Xiao TS and Stutz A: Activation and regulation of the inflammasomes. Nat Rev Immunol. 13:397–411. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, et al: Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 36:401–414. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Cabral A, Cabral JE, Wang A, Zhang Y, Liang H, Nikbakht D, Corona L, Hoffman HM and McNulty R: Differential binding of NLRP3 to non-oxidized and Ox-mtDNA mediates NLRP3 inflammasome activation. Commun Biol. 6:5782023. View Article : Google Scholar : PubMed/NCBI

45 

Zamyatina A and Heine H: Lipopolysaccharide recognition in the crossroads of TLR4 and caspase-4/11 mediated inflammatory pathways. Front Immunol. 11:5851462020. View Article : Google Scholar : PubMed/NCBI

46 

Rathinam VAK, Zhao Y and Shao F: Innate immunity to intracellular LPS. Nat Immunol. 20:527–533. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Downs KP, Nguyen H, Dorfleutner A and Stehlik C: An overview of the non-canonical inflammasome. Mol Aspects Med. 76:1009242020. View Article : Google Scholar : PubMed/NCBI

48 

Naseer N, Zhang J, Bauer R, Constant DA, Nice TJ, Brodsky IE, Rauch I and Shin S: Salmonella enterica serovar typhimurium induces NAIP/NLRC4- and NLRP3/ASC-independent, caspase-4-dependent inflammasome activation in human intestinal epithelial cells. Infect Immun. 90:e00663212022. View Article : Google Scholar : PubMed/NCBI

49 

Baker PJ, Boucher D, Bierschenk D, Tebartz C, Whitney PG, D'Silva DB, Tanzer MC, Monteleone M, Robertson AA, Cooper MA, et al: NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur J Immunol. 45:2918–2926. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Schmid-Burgk JL, Gaidt MM, Schmidt T, Ebert TS, Bartok E and Hornung V: Caspase-4 mediates non-canonical activation of the NLRP3 inflammasome in human myeloid cells. Eur J Immunol. 45:2911–2917. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Franchi L, Eigenbrod T and Núñez G: Cutting edge: TNF-alpha mediates sensitization to ATP and silica via the NLRP3 inflammasome in the absence of microbial stimulation. J Immunol. 183:792–796. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, et al: Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 183:787–791. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Chen X, Liu G, Yuan Y, Wu G, Wang S and Yuan L: NEK7 interacts with NLRP3 to modulate the pyroptosis in inflammatory bowel disease via NF-κB signaling. Cell Death Dis. 10:9062019. View Article : Google Scholar : PubMed/NCBI

54 

Liu G, Chen X, Wang Q and Yuan L: NEK7: A potential therapy target for NLRP3-related diseases. Biosci Trends. 14:74–82. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Sharif H, Wang L, Wang WL, Magupalli VG, Andreeva L, Qiao Q, Hauenstein AV, Wu Z, Núñez G, Mao Y and Wu H: Structural mechanism for NEK7-licensed activation of NLRP3 inflammasome. Nature. 570:338–343. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F and Alnemri ES: Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. J Biol Chem. 287:36617–36622. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Py BF, Kim MS, Vakifahmetoglu-Norberg H and Yuan J: Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol Cell. 49:331–338. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Lu B, Nakamura T, Inouye K, Li J, Tang Y, Lundbäck P, Valdes-Ferrer SI, Olofsson PS, Kalb T, Roth J, et al: Novel role of PKR in inflammasome activation and HMGB1 release. Nature. 488:670–674. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Li Y, Lin X, Wang W, Wang W, Cheng S, Huang Y, Zou Y, Ke J and Zhu L: The proinflammatory role of guanylate-binding protein 5 in inflammatory bowel diseases. Front Microbiol. 13:9269152022. View Article : Google Scholar : PubMed/NCBI

60 

Yang CS, Kim JJ, Kim TS, Lee PY, Kim SY, Lee HM, Shin DM, Nguyen LT, Lee MS, Jin HS, et al: Small heterodimer partner interacts with NLRP3 and negatively regulates activation of the NLRP3 inflammasome. Nat Commun. 6:61152015. View Article : Google Scholar : PubMed/NCBI

61 

Hara H, Tsuchiya K, Kawamura I, Fang R, Hernandez-Cuellar E, Shen Y, Mizuguchi J, Schweighoffer E, Tybulewicz V and Mitsuyama M: Phosphorylation of the adaptor ASC acts as a molecular switch that controls the formation of speck-like aggregates and inflammasome activity. Nat Immunol. 14:1247–1255. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Okada M, Matsuzawa A, Yoshimura A and Ichijo H: The lysosome rupture-activated TAK1-JNK pathway regulates NLRP3 inflammasome activation. J Biol Chem. 289:32926–32936. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Humke EW, Shriver SK, Starovasnik MA, Fairbrother WJ and Dixit VM: ICEBERG: A novel inhibitor of interleukin-1beta generation. Cell. 103:99–111. 2000. View Article : Google Scholar : PubMed/NCBI

64 

Druilhe A, Srinivasula SM, Razmara M, Ahmad M and Alnemri ES: Regulation of IL-1beta generation by Pseudo-ICE and ICEBERG, two dominant negative caspase recruitment domain proteins. Cell Death Differ. 8:649–657. 2001. View Article : Google Scholar : PubMed/NCBI

65 

Lamkanfi M, Denecker G, Kalai M, D'hondt K, Meeus A, Declercq W, Saelens X and Vandenabeele P: INCA, a novel human caspase recruitment domain protein that inhibits interleukin-1beta generation. J Biol Chem. 279:51729–51738. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Saleh M, Mathison JC, Wolinski MK, Bensinger SJ, Fitzgerald P, Droin N, Ulevitch RJ, Green DR and Nicholson DW: Enhanced bacterial clearance and sepsis resistance in caspase-12-deficient mice. Nature. 440:1064–1068. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Bedoya F, Sandler LL and Harton JA: Pyrin-only protein 2 modulates NF-kappaB and disrupts ASC:CLR interactions. J Immunol. 178:3837–3845. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Dorfleutner A, Bryan NB, Talbott SJ, Funya KN, Rellick SL, Reed JC, Shi X, Rojanasakul Y, Flynn DC and Stehlik C: Cellular pyrin domain-only protein 2 is a candidate regulator of inflammasome activation. Infect Immun. 75:1484–1492. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Khare S, Ratsimandresy RA, de Almeida L, Cuda CM, Rellick SL, Misharin AV, Wallin MC, Gangopadhyay A, Forte E, Gottwein E, et al: The PYRIN domain-only protein POP3 inhibits ALR inflammasomes and regulates responses to infection with DNA viruses. Nat Immunol. 15:343–353. 2014. View Article : Google Scholar : PubMed/NCBI

70 

de Almeida L, Khare S, Misharin AV, Patel R, Ratsimandresy RA, Wallin MC, Perlman H, Greaves DR, Hoffman HM, Dorfleutner A and Stehlik C: The PYRIN domain-only protein POP1 inhibits inflammasome assembly and ameliorates inflammatory disease. Immunity. 43:264–276. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Martin BN, Wang C, Willette-Brown J, Herjan T, Gulen MF, Zhou H, Bulek K, Franchi L, Sato T, Alnemri ES, et al: IKKα negatively regulates ASC-dependent inflammasome activation. Nat Commun. 5:49772014. View Article : Google Scholar : PubMed/NCBI

72 

Ito M, Yanagi Y and Ichinohe T: Encephalomyocarditis virus viroporin 2B activates NLRP3 inflammasome. PLoS Pathog. 8:e10028572012. View Article : Google Scholar : PubMed/NCBI

73 

Ermler ME, Traylor Z, Patel K, Schattgen SA, Vanaja SK, Fitzgerald KA and Hise AG: Rift Valley fever virus infection induces activation of the NLRP3 inflammasome. Virology. 449:174–180. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Pinar A, Dowling JK, Bitto NJ, Robertson AA, Latz E, Stewart CR, Drummond GR, Cooper MA, McAuley JL, Tate MD and Mansell A: PB1-F2 peptide derived from avian influenza A virus H7N9 induces inflammation via activation of the NLRP3 inflammasome. J Biol Chem. 292:826–836. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Xu J, Zhou Z, Zheng Y, Yang S, Huang K and Li H: Roles of inflammasomes in viral myocarditis. Front Cell Infect Microbiol. 13:11499112023. View Article : Google Scholar : PubMed/NCBI

76 

Wang W, Li G, De Wu, Luo Z, Pan P, Tian M, Wang Y, Xiao F, Li A, Wu K, et al: Zika virus infection induces host inflammatory responses by facilitating NLRP3 inflammasome assembly and interleukin-1β secretion. Nat Commun. 9:1062018. View Article : Google Scholar : PubMed/NCBI

77 

de Castro-Jorge LA, de Carvalho RVH, Klein TM, Hiroki CH, Lopes AH, Guimarães RM, Fumagalli MJ, Floriano VG, Agostinho MR, Slhessarenko RD, et al: The NLRP3 inflammasome is involved with the pathogenesis of Mayaro virus. PLoS Pathog. 15:e10079342019. View Article : Google Scholar : PubMed/NCBI

78 

Shrivastava G, Visoso-Carvajal G, Garcia-Cordero J, Leon-Juarez M, Chavez-Munguia B, Lopez T, Nava P, Villegas-Sepulveda N and Cedillo-Barron L: Dengue virus serotype 2 and its non-structural proteins 2A and 2B activate NLRP3 inflammasome. Front Immunol. 11:3522020. View Article : Google Scholar : PubMed/NCBI

79 

Xie WH, Ding J, Xie XX, Yang XH, Wu XF, Chen ZX, Guo QL, Gao WY, Wang XZ and Li D: Hepatitis B virus X protein promotes liver cell pyroptosis under oxidative stress through NLRP3 inflammasome activation. Inflamm Res. 69:683–696. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Zhi X, Zhang Y, Sun S, Zhang Z, Dong H, Luo X, Wei Y, Lu Z, Dou Y, Wu R, et al: NLRP3 inflammasome activation by Foot-and-mouth disease virus infection mainly induced by viral RNA and non-structural protein 2B. RNA Biol. 17:335–349. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Zhao C and Zhao W: NLRP3 inflammasome-A key player in antiviral responses. Front Immunol. 11:2112020. View Article : Google Scholar : PubMed/NCBI

82 

Harris J and Borg NA: The multifaceted roles of NLRP3-modulating proteins in virus infection. Front Immunol. 13:9874532022. View Article : Google Scholar : PubMed/NCBI

83 

Chen IY, Moriyama M, Chang MF and Ichinohe T: Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol. 10:502019. View Article : Google Scholar : PubMed/NCBI

84 

Guo HC, Jin Y, Zhi XY, Yan D and Sun SQ: NLRP3 inflammasome activation by viroporins of animal viruses. Viruses. 7:3380–3391. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Freeman TL and Swartz TH: Targeting the NLRP3 inflammasome in Severe COVID-19. Front Immunol. 11:15182020. View Article : Google Scholar : PubMed/NCBI

86 

da Costa LS, Outlioua A, Anginot A, Akarid K and Arnoult D: RNA viruses promote activation of the NLRP3 inflammasome through cytopathogenic effect-induced potassium efflux. Cell Death Dis. 10:3462019. View Article : Google Scholar : PubMed/NCBI

87 

Guo S, Yang C, Diao B, Huang X, Jin M, Chen L, Yan W, Ning Q, Zheng L, Wu Y and Chen Y: The NLRP3 inflammasome and IL-1β accelerate immunologically mediated pathology in experimental viral fulminant hepatitis. PLoS Pathog. 11:e10051552015. View Article : Google Scholar : PubMed/NCBI

88 

Guo H, Gao J, Taxman DJ, Ting JPY and Su L: HIV-1 infection induces interleukin-1β production via TLR8 protein-dependent and NLRP3 inflammasome mechanisms in human monocytes. J Biol Chem. 289:21716–21726. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Shil NK, Pokharel SM, Banerjee AK, Hoffman M and Bose S: Inflammasome antagonism by human parainfluenza virus type 3 c protein. J Virol. 92:e01776–17. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Moriyama M, Nagai M, Maruzuru Y, Koshiba T, Kawaguchi Y and Ichinohe T: Influenza virus-induced oxidized DNA activates inflammasomes. iScience. 23:1012702020. View Article : Google Scholar : PubMed/NCBI

91 

Barlan AU, Danthi P and Wiethoff CM: Lysosomal localization and mechanism of membrane penetration influence nonenveloped virus activation of the NLRP3 inflammasome. Virology. 412:306–314. 2011. View Article : Google Scholar : PubMed/NCBI

92 

Chakrabarti A, Banerjee S, Franchi L, Loo YM, Gale M Jr, Núñez G and Silverman RH: RNase L activates the NLRP3 inflammasome during viral infections. Cell Host Microbe. 17:466–477. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Subramanian N, Natarajan K, Clatworthy MR, Wang Z and Germain RN: The adaptor MAVS promotes NLRP3 mitochondrial localization and inflammasome activation. Cell. 153:348–361. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Li J, Hu L, Liu Y, Huang L, Mu Y, Cai X and Weng C: DDX19A senses viral RNA and mediates NLRP3-dependent inflammasome activation. J Immunol. 195:5732–5749. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Wang X, Jiang W, Yan Y, Gong T, Han J, Tian Z and Zhou R: RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway. Nat Immunol. 15:1126–1133. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Komune N, Ichinohe T, Ito M and Yanagi Y: Measles virus V protein inhibits NLRP3 inflammasome-mediated interleukin-1β secretion. J Virol. 85:13019–13026. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Komatsu T, Tanaka Y, Kitagawa Y, Koide N, Naiki Y, Morita N, Gotoh B and Yokochi T: Sendai virus V protein inhibits the secretion of interleukin-1β by preventing NLRP3 inflammasome assembly. J Virol. 92:e00842–18. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Pothlichet J, Meunier I, Davis BK, Ting JP, Skamene E, von Messling V and Vidal SM: Type I IFN triggers RIG-I/TLR3/NLRP3-dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog. 9:e10032562013. View Article : Google Scholar : PubMed/NCBI

99 

Park HS, Liu G, Thulasi Raman SN, Landreth SL, Liu Q and Zhou Y: NS1 protein of 2009 pandemic influenza A virus inhibits porcine NLRP3 inflammasome-mediated interleukin-1 beta production by suppressing ASC ubiquitination. J Virol. 92:e00022–18. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Cheung PHH, Ye ZW, Lee TWT, Chen H, Chan CP and Jin DY: PB1-F2 protein of highly pathogenic influenza A (H7N9) virus selectively suppresses RNA-induced NLRP3 inflammasome activation through inhibition of MAVS-NLRP3 interaction. J Leukoc Biol. 108:1655–1663. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Darweesh M, Kamel W, Gavrilin MA, Akusjärvi G and Svensson C: Adenovirus VA RNAI blocks ASC oligomerization and inhibits NLRP3 inflammasome activation. Front Immunol. 10:27912019. View Article : Google Scholar : PubMed/NCBI

102 

Gim E, Shim DW, Hwang I, Shin OS and Yu JW: Zika virus impairs host NLRP3-mediated inflammasome activation in an NS3-dependent manner. Immune Netw. 19:e402019. View Article : Google Scholar : PubMed/NCBI

103 

Haneklaus M, Gerlic M, Kurowska-Stolarska M, Rainey AA, Pich D, McInnes IB, Hammerschmidt W, O'Neill LA and Masters SL: Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1β production. J Immunol. 189:3795–3799. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Wang H, Lei X, Xiao X, Yang C, Lu W, Huang Z, Leng Q, Jin Q, He B, Meng G and Wang J: Reciprocal regulation between enterovirus 71 and the NLRP3 inflammasome. Cell Rep. 12:42–48. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Yu X, Lan P, Hou X, Han Q, Lu N, Li T, Jiao C, Zhang J, Zhang C and Tian Z: HBV inhibits LPS-induced NLRP3 inflammasome activation and IL-1β production via suppressing the NF-κB pathway and ROS production. J Hepatol. 66:693–702. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Johnson KE, Chikoti L and Chandran B: Herpes simplex virus 1 infection induces activation and subsequent inhibition of the IFI16 and NLRP3 inflammasomes. J Virol. 87:5005–5018. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2023
Volume 28 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng Q, Hua C, Liang Q and Cheng H: The NLRP3 inflammasome in viral infection (Review). Mol Med Rep 28: 160, 2023.
APA
Zheng, Q., Hua, C., Liang, Q., & Cheng, H. (2023). The NLRP3 inflammasome in viral infection (Review). Molecular Medicine Reports, 28, 160. https://doi.org/10.3892/mmr.2023.13047
MLA
Zheng, Q., Hua, C., Liang, Q., Cheng, H."The NLRP3 inflammasome in viral infection (Review)". Molecular Medicine Reports 28.3 (2023): 160.
Chicago
Zheng, Q., Hua, C., Liang, Q., Cheng, H."The NLRP3 inflammasome in viral infection (Review)". Molecular Medicine Reports 28, no. 3 (2023): 160. https://doi.org/10.3892/mmr.2023.13047