NLRP1 Haplotypes Associated with Vitiligo and Autoimmunity Increase Interleukin-1Β Processing Via the NLRP1 Inflammasome

Total Page:16

File Type:pdf, Size:1020Kb

NLRP1 Haplotypes Associated with Vitiligo and Autoimmunity Increase Interleukin-1Β Processing Via the NLRP1 Inflammasome NLRP1 haplotypes associated with vitiligo and autoimmunity increase interleukin-1β processing via the NLRP1 inflammasome Cecilia B. Levandowskia, Christina M. Maillouxa, Tracey M. Ferraraa, Katherine Gowana, Songtao Bena, Ying Jina,b, Kimberly K. McFannc, Paulene J. Hollanda, Pamela R. Faina,b,d, Charles A. Dinarelloe,1, and Richard A. Spritza,b,1 aHuman Medical Genetics and Genomics Program and bDepartment of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045; cColorado Biostatistics Consortium, Colorado School of Public Health, Aurora, CO 80045; and dBarbara Davis Center for Childhood Diabetes, and eDepartment of Medicine, University of Colorado School of Medicine, Aurora, CO 80045 Contributed by Charles A. Dinarello, January 8, 2013 (sent for review December 26, 2012) Nuclear localization leucine-rich-repeat protein 1 (NLRP1) is a key associated molecular patterns stimulate surface Toll-like recep- regulator of the innate immune system, particularly in the skin tors (TLRs) with subsequent assembly of the NLRP1 inflam- where, in response to molecular triggers such as pathogen- masome and activation of caspase-1. Active caspase-1 then associated or damage-associated molecular patterns, the NLRP1 cleaves the inactive IL-1β precursor (pro–IL-1β) to the mature inflammasome promotes caspase-1–dependent processing of bio- bioactive IL-1β (23), thereby stimulating downstream inflam- active interleukin-1β (IL-1β), resulting in IL-1β secretion and down- matory responses (23, 24). Because of its highly inflammatory stream inflammatory responses. NLRP1 is genetically associated and immunostimulating properties, the processing and release of with risk of several autoimmune diseases including generalized bioactive IL-1β are tightly controlled. However, in cryopyrin- vitiligo, Addison disease, type 1 diabetes, rheumatoid arthritis, associated periodic syndromes (CAPSs), which include familial and others. Here we identify a repertoire of variation in NLRP1 cold autoinflammatory disease, Muckle–Wells syndrome, and by deep DNA resequencing. Predicted functional variations in neonatal onset multi-inflammatory disease, amino acid sub- NLRP1 reside in several common high-risk haplotypes that differ stitutions in the NACHT domain of NLRP3 result in constitutive from the reference by multiple nonsynonymous substitutions. The activation of caspase-1 and processing and secretion of bioactive haplotypes that are high risk for disease share two substitutions, IL-1β, accompanied by marked systemic and local inflammation L155H and M1184V, and are inherited largely intact due to exten- (21, 25–28). Blood monocytes from affected patients release sive linkage disequilibrium across the region. Functionally, we significantly more mature bioactive IL-1β than cells from healthy found that peripheral blood monocytes from healthy subjects ho- individuals (28). Similarly, a dominant activating mutation in mozygous for the predominant high-risk haplotype 2A processed mouse Nlrp1a has recently been shown to result in a severe sys- significantly greater (P < 0.0001) amounts of the IL-1β precursor temic inflammatory phenotype associated with greatly elevated to mature bioactive IL-1β under basal (resting) conditions and in release of active IL-1β (29). response to Toll-like receptor (TLR) agonists (TLR2 and TLR4) com- In the present study we have investigated the hypothesis that pared with monocytes from subjects homozygous for the refer- common inherited variation in human NLRP1, which is associ- ence haplotype 1. The increase in basal release was 1.8-fold ated with genetic risk of autoimmune disease, similarly results in greater in haplotype 2A monocytes, and these differences be- constitutively increased processing and release of bioactive IL-1β. tween the two haplotypes were consistently observed three times By next-generation DNA sequencing, we identified common over a 3-mo period; no differences were observed for IL-1α or NLRP1 haplotypes containing multiple predicted functional var- TNFα. NLRP1 RNA and protein levels were not altered by the pre- iants that are associated with disease. We next studied the in vitro dominant high-risk haplotype, indicating that altered function of production of cytokines from monocytes of healthy individuals the corresponding multivariant NLRP1 polypeptide predisposes to homozygous for the most prevalent high-risk NLRP1 haplotype autoimmune diseases by activation of the NLRP1 inflammasome. and compared the responses to monocytes from subjects with the low-risk reference haplotype. Our findings indicate that these cytokine | DNA sequencing | interleukin-1beta multivariant NLRP1 haplotypes predispose to common autoim- mune diseases by activation of the NLRP1 inflammasome. utoimmune diseases are a diverse group of more than 80 Achronic disorders in which the immune system attacks “self” Results tissues and cells (1), altogether affecting 3–5% of the U.S. pop- NLRP1 Sequencing Identifies Conserved Disease-Associated Haplotypes ulation (2). Concomitant occurrence of multiple autoimmune That Differ by Multiple Nonsynonymous Substitutions. To identify diseases in some patients is well known (3–5) and results from genetic variation in NLRP1 that might contribute to autoimmune shared genetic and perhaps environmental risk factors among susceptibility, we resequenced the complete NLRP1 gene region these different diseases (6–8). Polymorphisms of the nuclear lo- in probands of the 114 families with multiple cases of vitiligo and calization leucine-rich-repeat protein 1 (NLRP1) gene encoding other autoimmune diseases in which we originally established the NACHT (NAIP, CIITA, HET-E, TP1) domain of NLRP1 genetic linkage and association with NLRP1 (9, 10). The 161.5 kb have been associated with a number of autoimmune diseases, including vitiligo (9, 10), Addison disease (11, 12), type 1 di- abetes (11), celiac disease (13), systemic lupus erythematosus Author contributions: P.R.F., C.A.D., and R.A.S. designed research; C.B.L., C.M.M., T.M.F., (14), rheumatoid arthritis (15), systemic sclerosis (16), Kawasaki S.B., and Y.J. performed research; C.M.M., T.M.F., and P.J.H. contributed new reagents/ fl analytic tools; C.B.L., C.M.M., T.M.F., K.G., S.B., Y.J., K.K.M., P.R.F., C.A.D., and R.A.S. disease (17), as well as steroid responsiveness in in ammatory analyzed data; and C.B.L., K.K.M., C.A.D., and R.A.S. wrote the paper. fi bowel disease (18), although the speci c biological mechanism The authors declare no conflict of interest. underlying these genetic associations remains unknown. 1To whom correspondence may be addressed. E-mail: [email protected] or richard. NLRP1 is a regulator of the innate immune response (19–21) [email protected]. and is expressed in many immunocompetent cell types, particu- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. larly the Langerhans cells of the skin (22). Pathogen- or damage- 1073/pnas.1222808110/-/DCSupplemental. 2952–2956 | PNAS | February 19, 2013 | vol. 110 | no. 8 www.pnas.org/cgi/doi/10.1073/pnas.1222808110 Downloaded by guest on September 23, 2021 region sequenced (chr17:5386564–5548062) included the entire otherwise specific to haplotype 3. In addition to these major 70.4 kb NLRP1 structural gene, as well as 60.2 kb upstream and multivariant haplotypes, we also observed four singleton non- 30.9 kb downstream. We observed a total of 656 sequence variants synonymous substitutions, G354D, E869K, T1113M, and R1289H, (Dataset S1), including 261 not annotated in the Short Genetic of which only T1113M cosegregated with autoimmunity in the Variations database (dbSNP), Build 135. Within the exons, these corresponding multiplex family. Of the 16 observed non- included 16 nonsynonymous variants, with no observed nonsense synonymous substitutions, bioinformatic analyses predict that variants, splice junction variants, or coding region indels. We six are probably or possibly deleterious (Fig. 1), although in- verified all previously unannotated nonsynonymous variants by dividual in silico functional predictions are of uncertain val- Sanger sequencing in the corresponding patients, and we in- idityinthecontextofsuchmultivariant haplotypes. Of these, tegrated the observed sequence variants with high-density NLRP1 L155H is the only predicted deleterious substitution shared by region SNP data in these same families (9). both high-risk haplotypes 2A and 3, which additionally share The NLRP1 region consists of three main haplotype blocks, the predicted benign variant M1184V. comprising the NLRP1 structural gene, the 5′-untranslated se- quence and extended promoter region, and the 3′-untranslated Increased Processing of Mature IL-1β from Monocytes of Healthy and 3′- flanking region. Of the 16 nonsynonymous variants, 12 Subjects with the Prevalent High-Risk NLRP1 Haplotype. To de- were genotyped directly or were sufficiently frequent to permit termine whether the multivariant NLRP1 polypeptides encoded accurate genotype imputation (r2 > 0.3) in the corresponding by disease-associated NLRP1 haplotypes differentially affect families. Previous family-based association analyses showed that NLRP1 function, we screened healthy individuals to identify rs12150220 (L155H) was the single variant most significantly subjects alternatively homozygous for the NLRP1 reference associated with vitiligo (9). However, the L155H variant occurs haplotype 1 or for the prevalent high-risk NLRP1 haplotype 2A. on three main haplotypes, each differing by multiple additional We genotyped a total of 115
Recommended publications
  • ATP-Binding and Hydrolysis in Inflammasome Activation
    molecules Review ATP-Binding and Hydrolysis in Inflammasome Activation Christina F. Sandall, Bjoern K. Ziehr and Justin A. MacDonald * Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; [email protected] (C.F.S.); [email protected] (B.K.Z.) * Correspondence: [email protected]; Tel.: +1-403-210-8433 Academic Editor: Massimo Bertinaria Received: 15 September 2020; Accepted: 3 October 2020; Published: 7 October 2020 Abstract: The prototypical model for NOD-like receptor (NLR) inflammasome assembly includes nucleotide-dependent activation of the NLR downstream of pathogen- or danger-associated molecular pattern (PAMP or DAMP) recognition, followed by nucleation of hetero-oligomeric platforms that lie upstream of inflammatory responses associated with innate immunity. As members of the STAND ATPases, the NLRs are generally thought to share a similar model of ATP-dependent activation and effect. However, recent observations have challenged this paradigm to reveal novel and complex biochemical processes to discern NLRs from other STAND proteins. In this review, we highlight past findings that identify the regulatory importance of conserved ATP-binding and hydrolysis motifs within the nucleotide-binding NACHT domain of NLRs and explore recent breakthroughs that generate connections between NLR protein structure and function. Indeed, newly deposited NLR structures for NLRC4 and NLRP3 have provided unique perspectives on the ATP-dependency of inflammasome activation. Novel molecular dynamic simulations of NLRP3 examined the active site of ADP- and ATP-bound models. The findings support distinctions in nucleotide-binding domain topology with occupancy of ATP or ADP that are in turn disseminated on to the global protein structure.
    [Show full text]
  • NLR Members in Inflammation-Associated
    Cellular & Molecular Immunology (2017) 14, 403–405 & 2017 CSI and USTC All rights reserved 2042-0226/17 $32.00 www.nature.com/cmi RESEARCH HIGHTLIGHT NLR members in inflammation-associated carcinogenesis Ha Zhu1,2 and Xuetao Cao1,2,3 Cellular & Molecular Immunology (2017) 14, 403–405; doi:10.1038/cmi.2017.14; published online 3 April 2017 hronic inflammation is regarded as an impor- nucleotide-binding and oligomerization domain IL-2,8 and NAIP was found to regulate the STAT3 Ctant factor in cancer progression. In addition (NOD)-like receptors (NLRs). TLRs and CLRs are pathway independent of inflammasome formation.9 to the immune surveillance function in the early located in the plasma membranes, whereas RLRs, The AOM/DSS model is the most popular model stage of tumorigenesis, inflammation is also known ALRs and NLRs are intracellular PRRs.3 Unlike used to study the function of NLRs in fl fl as one of the hallmarks of cancer and can supply other families that have been shown to bind their in ammation-associated carcinogenesis. In amma- the tumor microenvironment with bioactive mole- specific cognate ligands, the distinct ligands for somes initiated by NLRs or AIM2 have been widely cules and favor the development of other hallmarks NLRs are still unknown. In fact, mounting evidence reported to participate in the maintenance of 10,11 Nlrp3 Nlrp6 of cancer, such as genetic instability and angiogen- suggests that NLRs function as cytoplasmic sensors intestinal homeostasis. -/-, -/-, Nlrc4 Nlrp1 Nlrx1 Nlrp12 esis. Moreover, inflammation contributes to the and participate in modulating TLR, RLR and CLR -/-, -/-, -/- and -/- mice are 4 more susceptible to AOM/DSS-induced colorectal changing tumor microenvironment by altering signaling pathways.
    [Show full text]
  • The NLRP3 and NLRP1 Inflammasomes Are Activated In
    Saresella et al. Molecular Neurodegeneration (2016) 11:23 DOI 10.1186/s13024-016-0088-1 RESEARCHARTICLE Open Access The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease Marina Saresella1*†, Francesca La Rosa1†, Federica Piancone1, Martina Zoppis1, Ivana Marventano1, Elena Calabrese1, Veronica Rainone2, Raffaello Nemni1,3, Roberta Mancuso1 and Mario Clerici1,3 Abstract Background: Interleukin-1 beta (IL-1β) and its key regulator, the inflammasome, are suspected to play a role in the neuroinflammation observed in Alzheimer’s disease (AD); no conclusive data are nevertheless available in AD patients. Results: mRNA for inflammasome components (NLRP1, NLRP3, PYCARD, caspase 1, 5 and 8) and downstream effectors (IL-1β, IL-18) was up-regulated in severe and MILD AD. Monocytes co-expressing NLRP3 with caspase 1 or caspase 8 were significantly increased in severe AD alone, whereas those co-expressing NLRP1 and NLRP3 with PYCARD were augmented in both severe and MILD AD. Activation of the NLRP1 and NLRP3 inflammasomes in AD was confirmed by confocal microscopy proteins co-localization and by the significantly higher amounts of the pro-inflammatory cytokines IL-1β and IL-18 being produced by monocytes. In MCI, the expression of NLRP3, but not the one of PYCARD or caspase 1 was increased, indicating that functional inflammasomes are not assembled in these individuals: this was confirmed by lack of co-localization and of proinflammatory cytokines production. Conclusions: The activation of at least two different inflammasome complexes
    [Show full text]
  • Complementary Regulation of Caspase-1 and IL-1Β Reveals Additional Mechanisms of Dampened Inflammation in Bats
    Complementary regulation of caspase-1 and IL-1β reveals additional mechanisms of dampened inflammation in bats Geraldine Goha,1, Matae Ahna,1, Feng Zhua, Lim Beng Leea, Dahai Luob,c, Aaron T. Irvinga,d,2, and Lin-Fa Wanga,e,2 aProgramme in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, 169857, Singapore; bLee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore; cNTU Institute of Structural Biology, Nanyang Technological University, 636921, Singapore; dZhejiang University–University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University International Campus, Haining, 314400, China; and eSinghealth Duke–NUS Global Health Institute, 169857, Singapore Edited by Vishva M. Dixit, Genentech, San Francisco, CA, and approved September 14, 2020 (received for review February 21, 2020) Bats have emerged as unique mammalian vectors harboring a experimental confirmation is rare. We recently demonstrated diverse range of highly lethal zoonotic viruses with minimal that NLRP3 is dampened in bats as a result of loss-of-function clinical disease. Despite having sustained complete genomic loss bat-specific isoforms and impaired transcriptional priming (9). of AIM2, regulation of the downstream inflammasome response in The stimulator of IFN genes (STING), a key adaptor to the bats is unknown. AIM2 sensing of cytoplasmic DNA triggers ASC DNA-sensing cGAS protein, is also exclusively mutated at S358 aggregation and recruits caspase-1, the central inflammasome ef- in bats, resulting in a reduced IFN response to HSV1 (10). We fector enzyme, triggering cleavage of cytokines such as IL-1β and previously reported a complete absence of Absent in melanoma inducing GSDMD-mediated pyroptotic cell death.
    [Show full text]
  • The Molecular Links Between Cell Death and Inflammasome
    cells Review The Molecular Links between Cell Death and Inflammasome Kwang-Ho Lee 1,2 and Tae-Bong Kang 1,2,* 1 Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea 2 Research Institute of Inflammatory Diseases, Konkuk University, Chungju 27478, Korea * Correspondence: [email protected]; Tel.: +82-43-840-3904; Fax: +82-43-852-3616 Received: 30 July 2019; Accepted: 9 September 2019; Published: 10 September 2019 Abstract: Programmed cell death pathways and inflammasome activation pathways can be genetically and functionally separated. Inflammasomes are specialized protein complexes that process pro-inflammatory cytokines, interleukin-1β (IL-1β), and IL-18 to bioactive forms for protection from a wide range of pathogens, as well as environmental and host-derived danger molecules. Programmed cell death has been extensively studied, and its role in the development, homeostasis, and control of infection and danger is widely appreciated. Apoptosis and the recently recognized necroptosis are the best-characterized forms of programmed death, and the interplay between them through death receptor signaling is also being studied. Moreover, growing evidence suggests that many of the signaling molecules known to regulate programmed cell death can also modulate inflammasome activation in a cell-intrinsic manner. Therefore, in this review, we will discuss the current knowledge concerning the role of the signaling molecules originally associated with programmed cell death in the activation of inflammasome and IL-1β processing. Keywords: inflammasome; apoptosis; necroptosis; programmed cell death; Caspase-8; RIPK1/3; MLKL; PGAM5; DRP1 1. Introduction Homeostasis is a principle property of living organisms and it is maintained at the systemic, tissue, and cellular levels through the homeostatic control system.
    [Show full text]
  • NAIP5/NLRC4 Inflammasomes Compounds Inhibit the NLRP1, NLRP3, and Arsenic Trioxide and Other Arsenical
    The Journal of Immunology Arsenic Trioxide and Other Arsenical Compounds Inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 Inflammasomes Nolan K. Maier,* Devorah Crown,* Jie Liu,† Stephen H. Leppla,* and Mahtab Moayeri* Inflammasomes are large cytoplasmic multiprotein complexes that activate caspase-1 in response to diverse intracellular danger signals. Inflammasome components termed nucleotide-binding oligomerization domain–like receptor (NLR) proteins act as sensors for pathogen-associated molecular patterns, stress, or danger stimuli. We discovered that arsenicals, including arsenic trioxide and sodium arsenite, inhibited activation of the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes by their respective activat- ing signals, anthrax lethal toxin, nigericin, and flagellin. These compounds prevented the autoproteolytic activation of caspase-1 and the processing and secretion of IL-1b from macrophages. Inhibition was independent of protein synthesis induction, proteasome-mediated protein breakdown, or kinase signaling pathways. Arsenic trioxide and sodium arsenite did not directly modify or inhibit the activity of preactivated recombinant caspase-1. Rather, they induced a cellular state inhibitory to both the autoproteolytic and substrate cleavage activities of caspase-1, which was reversed by the reactive oxygen species scavenger N-acetylcysteine but not by reducing agents or NO pathway inhibitors. Arsenicals provided protection against NLRP1-dependent anthrax lethal toxin–mediated cell death and prevented NLRP3-dependent neutrophil recruitment in a monosodium urate crystal inflammatory murine peritonitis model. These findings suggest a novel role in inhibition of the innate immune response for arsenical compounds that have been used as therapeutics for a few hundred years. The Journal of Immunology, 2014, 192: 763–770. nflammasomes are large cytoplasmic multiprotein complexes domain–containing protein 4 (NLRC4) inflammasome by direct that form in response to intracellular danger signals.
    [Show full text]
  • Evasion of Inflammasome Activation by Microbial Pathogens
    Evasion of inflammasome activation by microbial pathogens Tyler K. Ulland, … , Polly J. Ferguson, Fayyaz S. Sutterwala J Clin Invest. 2015;125(2):469-477. https://doi.org/10.1172/JCI75254. Review Activation of the inflammasome occurs in response to infection with a wide array of pathogenic microbes. The inflammasome serves as a platform to activate caspase-1, which results in the subsequent processing and secretion of the proinflammatory cytokines IL-1β and IL-18 and the initiation of an inflammatory cell death pathway termed pyroptosis. Effective inflammasome activation is essential in controlling pathogen replication as well as initiating adaptive immune responses against the offending pathogens. However, a number of pathogens have developed strategies to evade inflammasome activation. In this Review, we discuss these pathogen evasion strategies as well as the potential infectious complications of therapeutic blockade of IL-1 pathways. Find the latest version: https://jci.me/75254/pdf The Journal of Clinical Investigation REVIEW Evasion of inflammasome activation by microbial pathogens Tyler K. Ulland,1,2 Polly J. Ferguson,3 and Fayyaz S. Sutterwala1,2,4,5 1Inflammation Program, 2Interdisciplinary Program in Molecular and Cellular Biology, 3Department of Pediatrics, and 4Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA. 5Veterans Affairs Medical Center, Iowa City, Iowa, USA. Activation of the inflammasome occurs in response to infection with a wide array of pathogenic microbes. The inflammasome serves as a platform to activate caspase-1, which results in the subsequent processing and secretion of the proinflammatory cytokines IL-1β and IL-18 and the initiation of an inflammatory cell death pathway termed pyroptosis.
    [Show full text]
  • NAIP5/NLRC4 Inflammasomes Compounds Inhibit the NLRP1
    Arsenic Trioxide and Other Arsenical Compounds Inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 Inflammasomes This information is current as Nolan K. Maier, Devorah Crown, Jie Liu, Stephen H. Leppla of September 26, 2021. and Mahtab Moayeri J Immunol 2014; 192:763-770; Prepublished online 13 December 2013; doi: 10.4049/jimmunol.1301434 http://www.jimmunol.org/content/192/2/763 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2013/12/13/jimmunol.130143 Material 4.DC1 http://www.jimmunol.org/ References This article cites 55 articles, 9 of which you can access for free at: http://www.jimmunol.org/content/192/2/763.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 26, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Arsenic Trioxide and Other Arsenical Compounds Inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 Inflammasomes Nolan K. Maier,* Devorah Crown,* Jie Liu,† Stephen H. Leppla,* and Mahtab Moayeri* Inflammasomes are large cytoplasmic multiprotein complexes that activate caspase-1 in response to diverse intracellular danger signals.
    [Show full text]
  • NOD-Like Receptors (Nlrs) and Inflammasomes
    International Edition www.adipogen.com NOD-like Receptors (NLRs) and Inflammasomes In mammals, germ-line encoded pattern recognition receptors (PRRs) detect the presence of pathogens through recognition of pathogen-associated molecular patterns (PAMPs) or endogenous danger signals through the sensing of danger-associated molecular patterns (DAMPs). The innate immune system comprises several classes of PRRs that allow the early detection of pathogens at the site of infection. The membrane-bound toll-like receptors (TLRs) and C-type lectin receptors (CTRs) detect PAMPs in extracellular milieu and endo- somal compartments. TRLs and CTRs cooperate with PRRs sensing the presence of cytosolic nucleic acids, like RNA-sensing RIG-I (retinoic acid-inducible gene I)-like receptors (RLRs; RLHs) or DNA-sensing AIM2, among others. Another set of intracellular sensing PRRs are the NOD-like receptors (NLRs; nucleotide-binding domain leucine-rich repeat containing receptors), which not only recognize PAMPs but also DAMPs. PAMPs FUNGI/PROTOZOA BACTERIA VIRUSES MOLECULES C. albicans A. hydrophila Adenovirus Bacillus anthracis lethal Plasmodium B. brevis Encephalomyo- toxin (LeTx) S. cerevisiae E. coli carditis virus Bacterial pore-forming L. monocytogenes Herpes simplex virus toxins L. pneumophila Influenza virus Cytosolic dsDNA N. gonorrhoeae Sendai virus P. aeruginosa Cytosolic flagellin S. aureus MDP S. flexneri meso-DAP S. hygroscopicus S. typhimurium DAMPs MOLECULES PARTICLES OTHERS DNA Uric acid UVB Extracellular ATP CPPD Mutations R837 Asbestos Cytosolic dsDNA Irritants Silica Glucose Alum Hyaluronan Amyloid-b Hemozoin Nanoparticles FIGURE 1: Overview on PAMPs and DAMPs recognized by NLRs. NOD-like Receptors [NLRs] The intracellular NLRs organize signaling complexes such as inflammasomes and NOD signalosomes.
    [Show full text]
  • Identification of the Pyroptosis‑Related Prognostic Gene Signature and the Associated Regulation Axis in Lung Adenocarcinoma
    www.nature.com/cddiscovery ARTICLE OPEN Identification of the pyroptosis‑related prognostic gene signature and the associated regulation axis in lung adenocarcinoma ✉ Wanli Lin1, Ying Chen1, Bomeng Wu1, Ying chen1 and Zuwei Li1 © The Author(s) 2021 Lung adenocarcinoma (LUAD) remains the most common deadly disease and has a poor prognosis. Pyroptosis could regulate tumour cell proliferation, invasion, and metastasis, thereby affecting the prognosis of cancer patients. However, the role of pyroptosis-related genes (PRGs) in LUAD remains unclear. In our study, comprehensive bioinformatics analysis was performed to construct a prognostic gene model and ceRNA network. The correlations between PRGs and tumour-immune infiltration, tumour mutation burden, and microsatellite instability were evaluated using Pearson’s correlation analysis. A total of 23 PRGs were upregulated or downregulated in LUAD. The genetic mutation variation landscape of PRG in LUAD was also summarised. Functional enrichment analysis revealed that these 33 PRGs were mainly involved in pyroptosis, the NOD-like receptor signalling pathway, and the Toll-like receptor signalling pathway. Prognosis analysis indicated a poor survival rate in LUAD patients with low expression of NLRP7, NLRP1, NLRP2, and NOD1 and high CASP6 expression. A prognostic PRG model constructed using the above five prognostic genes could predict the overall survival of LUAD patients with medium-to-high accuracy. Significant correlation was observed between prognostic PRGs and immune-cell infiltration, tumour mutation burden, and microsatellite instability. A ceRNA network was constructed to identify a lncRNA KCNQ1OT1/miR-335-5p/NLRP1/NLRP7 regulatory axis in LUAD. In conclusion, we performed a comprehensive bioinformatics analysis and identified a prognostic PRG signature containing five genes (NLRP7, NLRP1, NLRP2, NOD1, and CASP6) for LUAD patients.
    [Show full text]
  • Inflammasomes Inhibits the NLRP1 and NLRP3 2 the Cyclopentenone Prostaglandin 15D-PGJ
    The Cyclopentenone Prostaglandin 15d-PGJ2 Inhibits the NLRP1 and NLRP3 Inflammasomes This information is current as Nolan K. Maier, Stephen H. Leppla and Mahtab Moayeri of October 1, 2021. J Immunol 2015; 194:2776-2785; Prepublished online 13 February 2015; doi: 10.4049/jimmunol.1401611 http://www.jimmunol.org/content/194/6/2776 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2015/02/13/jimmunol.140161 Material 1.DCSupplemental References This article cites 69 articles, 25 of which you can access for free at: http://www.jimmunol.org/ http://www.jimmunol.org/content/194/6/2776.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on October 1, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology The Cyclopentenone Prostaglandin 15d-PGJ2 Inhibits the NLRP1 and NLRP3 Inflammasomes Nolan K. Maier, Stephen H. Leppla, and Mahtab Moayeri Inflammasomes are cytosolic protein complexes that respond to diverse danger signals by activating caspase-1.
    [Show full text]
  • Neurovascular Inflammaging in Health and Disease
    cells Review Neurovascular Inflammaging in Health and Disease Ádám Mészáros 1,2 , Kinga Molnár 1,3, Bernát Nógrádi 1,4 , Zsófia Hernádi 1,4, Ádám Nyúl-Tóth 1,5, Imola Wilhelm 1,6 and István A. Krizbai 1,6,* 1 Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; [email protected] (Á.M.); [email protected] (K.M.); [email protected] (B.N.); zsofi[email protected] (Z.H.); [email protected] (Á.N.-T.); [email protected] (I.W.) 2 Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary 3 Theoretical Medicine Doctoral School, University of Szeged, 6720 Szeged, Hungary 4 Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary 5 Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA 6 Institute of Life Sciences, Vasile Goldi¸sWestern University of Arad, 310414 Arad, Romania * Correspondence: [email protected]; Tel.: +36-62-599-794 Received: 22 June 2020; Accepted: 2 July 2020; Published: 4 July 2020 Abstract: Aging is characterized by a chronic low-grade sterile inflammation dubbed as inflammaging, which in part originates from accumulating cellular debris. These, acting as danger signals with many intrinsic factors such as cytokines, are sensed by a network of pattern recognition receptors and other cognate receptors, leading to the activation of inflammasomes. Due to the inflammasome activity-dependent increase in the levels of pro-inflammatory interleukins (IL-1β, IL-18), inflammation is initiated, resulting in tissue injury in various organs, the brain and the spinal cord included.
    [Show full text]