NAIP5/NLRC4 Inflammasomes Compounds Inhibit the NLRP1, NLRP3, and Arsenic Trioxide and Other Arsenical

Total Page:16

File Type:pdf, Size:1020Kb

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. These binding (5, 6). The exact mechanisms by which many disparate I diverse danger signals include pathogen-derived stimuli such signals activate the “promiscuous” NLRP3 inflammasome are un- as bacterial toxins, flagellin, and dsDNA; self-derived molecules known (2). The end result of activation of all inflammasome sensors such as uric acid, amyloid crystals, cholesterol, and ATP; and sig- is the recruitment of caspase-1 to the sensor complex, followed by nals of environmental origin such as aluminum hydroxide, asbestos, its autoproteolytic activation. Activated caspase-1 then rapidly and UV radiation (for reviews, see Refs. 1, 2). The nucleotide- processes the proinflammatory cytokines IL-1b and IL-18 to mature binding oligomerization domain–like receptor leucine-rich repeat forms, allowing their secretion. These cytokines, which are the first proteins (NLRPs), which act as the sensor components of inflam- line of defense for the innate immune response, initiate a cascade of masomes, are activated by several mechanisms. For example, an- other immunological responses. Inflammasome activation is often thrax lethal toxin (LT), a bipartite toxin made of a receptor binding accompanied by a caspase-1–dependent rapid cell death known as moiety (protective Ag [PA]) and a protease (lethal factor [LF]), pyroptosis (for reviews, see Refs. 1, 2). activates rodent NLRP1 inflammasomes by cleaving them in an N- Not surprisingly, inflammasomes and the innate immune response terminal domain (3, 4). Flagellin activates the NLR family apoptosis play a key role in many infections (7). However, the proinflammatory inhibitory protein 5 (NAIP5)/NLR family caspase-1 recruitment response initiated by inflammasomes has also been implicated in metabolic disorders such as diabetes and inflammatory diseases such as gout and arthritis (8). Furthermore, polymorphisms in the *Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Dis- inflammasome NLR sensors are associated with diseases including eases, National Institutes of Health, Bethesda, MD 20892; and †Center for Molecular vitiligo, rheumatoid arthritis, and Alzheimer’s disease (1). The Medicine, National Heart Lung and Blood Institute, National Institutes of Health, chronic inflammation etiologically associated with numerous can- Bethesda, MD 20892 cers, most notably gastric, hepatic, and colorectal cancers, has also Received for publication May 31, 2013. Accepted for publication November 12, 2013. been linked to activation of these sensors (9). Thus, the role played This work was supported in part by the Intramural Research Program of the National by inflammasome-initiated inflammation in human disease has led Institute of Allergy and Infectious Diseases. to much interest in developing therapeutics targeting inflammasomes Address correspondence and reprint requests to Dr. Mahtab Moayeri, National In- or caspase-1. stitutes of Health Building 33, Room 1W20B, Bethesda, MD 20892. E-mail address: In this study, we show that activation of multiple inflammasomes [email protected] is inhibited by arsenical compounds. Sodium arsenite (NaAsO2) The online version of this article contains supplemental material. and arsenic trioxide (As2O3-Trisenox, a drug with established clinical Abbreviations used in this article: APL, acute promyelocytic leukemia; As2O3, arse- efficacy in treating a number of hematological cancers, including nic trioxide; BMDM, bone marrow–derived macrophage; Boc-D-CMK, Boc-Asp (OBzl)-chloromethylketone; FlaTox, toxin consisting of fusion protein between lethal acute promyelocytic leukemia (APL) and multiple myeloma (10), factor N terminus and flagellin and protective Ag; LF, lethal factor; LFn-Fla, fusion inhibit LT-induced inflammasome-dependent macrophage pyrop- protein between lethal factor N terminus and flagellin; LT, lethal toxin (toxin con- tosis when used at clinically relevant doses. These compounds not sisting of lethal factor and protective Ag); MSU, monosodium urate; NaAsO2, so- dium arsenite; NAC, N-acetylcysteine; NAIP5, NLR family apoptosis inhibitory only inhibit NLRP1 inflammasome activation by LT, but also the protein 5; NLR, nucleotide-binding oligomerization domain–like receptor; NLRC4, NAIP5/NLRC4 and NLRP3 inflammasome responses to their effec- NLR family caspase-1 recruitment domain–containing protein 4; NLRP, nucleotide- binding oligomerization domain–like receptor, leucine-rich repeat protein; PA, pro- tors. We found that arsenical compounds inhibit both caspase-1 tective Ag; PML, promyelocytic leukemia protein; ROS, reactive oxygen species. self-activating autoproteolytic activity as well as preactivated www.jimmunol.org/cgi/doi/10.4049/jimmunol.1301434 764 ARSENIC COMPOUNDS INHIBIT THE INFLAMMASOME recombinant caspase-1. The inhibition does not occur through or times (as described in the figure legends). Cells were then treated with direct modification or inhibition of caspase-1 enzymatic function, LT, FlaTox, or medium. Cell viability was assessed by MTT (Sigma-Aldrich) but rather through induction of a cytoplasmic environment in intact as previously described (14). In selected flow cytometry experiments, propidium iodide was used for viability analysis on a BD LSR II flow cells that is inhibitory to its activity. Our findings suggest a novel cytometer (BD Biosciences). role for arsenical compounds as inflammasome inhibitors, with possible off-target utility for treatment of inflammatory conditions, MEK, caspase-1, and IL-1b cleavage as well as a possible explanation of the mechanism for As2O3 ef- RAW264.7 and BALB/cJ BMDM cells were treated with LPS (1 mg/ml) for ficacy in cytokine-dependent hematological cancers. 2 h, with or without drugs (at doses and timing indicated in the figure legends), prior to addition of inflammasome activators (LT, FlaTox, or nigericin, at indicated doses). Cells were then lysed and processed for Materials and Methods Western blotting using primary Abs as previously described (14) in con- Reagents junction with infrared dye–conjugated secondary Abs and visualization with the Odyssey infrared imaging system (LI-COR Biosciences). As2O3 and arsenic (III) chloride were purchased from Alfa Aesar (Ward Hill, MA). Other arsenicals included sodium arsenate (MP Biomedicals, Solon, In vitro caspase-1 assay OH) and arsenic (V) oxide (Strem Chemicals, Newburyport, MA). Cacodylic acid, cycloheximide, actinomycin D, puromycin, buthionine sulfoximine, LPS (50 ng/ml for 8 h, or 1 mg/ml for 2 h) was used to induce IL-1b as N-acetylcysteine (NAC), uric acid, and propidium iodide were from Sigma- a substrate for recombinant caspase-1. Sucrose buffer (250 mM sucrose, Aldrich (St. Louis, MO). Sodium fluoride, sodium orthovanadate, and 10 mM HEPES) lysates of LPS-treated BALB/cJ BMDMs or RAW264.7 NaAsO2 were obtained from Fisher Scientific (Pittsburgh, PA). Staurosporine cells were incubated with 1 U active recombinant mouse caspase-1 per 50 ml was from Biotium (Hayward, CA). Nigericin, anti-Mek1 NT Ab (444942), lysate (MBL International, Woburn, MA) in the presence or absence of G lactacystin, N -monomethyl-L-arginine, and ultrapure LPS were purchased arsenical drug, Boc-Asp(OBzl)-chloromethylketone (Boc-D-CMK; Anas- from Calbiochem (San Diego, CA). Anti-Mek3 NT Ab (sc-959), anti-actin pec, San Jose, CA), or reducing agent for 3 h at 37˚C. In other experiments (sc-1616), and anti-caspase1 p10 Ab (sc-514) were from Santa Cruz Bio- cells were first pretreated with arsenical drugs for 0.5–8 h (as indicated in technology (Santa Cruz, CA). Alexa Fluor 488–conjugated anti-Ly6 Ab was the
Recommended publications
  • Scholarly Commons NLRX1 Modulates Differentially NLRP3
    University of the Pacific Scholarly Commons Dugoni School of Dentistry Faculty Articles Arthur A. Dugoni School of Dentistry 10-1-2018 NLRX1 modulates differentially NLRP3 inflammasome activation and NF-κB signaling during Fusobacterium nucleatum infection Shu Chen Hung University of the Pacific Arthur A. Dugoni School of Dentistry Pei Rong Huang Chang Gung University Cassio Luiz Coutinho Almeida-Da-Silva University of the Pacific Arthur A. Dugoni School of Dentistry, [email protected] Kalina R. Atanasova University of Florida Ozlem Yilmaz Medical University of South Carolina See next page for additional authors Follow this and additional works at: https://scholarlycommons.pacific.edu/dugoni-facarticles Part of the Dentistry Commons Recommended Citation Hung, S., Huang, P., Almeida-Da-Silva, C. L., Atanasova, K. R., Yilmaz, O., & Ojcius, D. M. (2018). NLRX1 modulates differentially NLRP3 inflammasome activation and NF-κB signaling during Fusobacterium nucleatum infection. Microbes and Infection, 20(9-10), 615–625. DOI: 10.1016/j.micinf.2017.09.014 https://scholarlycommons.pacific.edu/dugoni-facarticles/705 This Article is brought to you for free and open access by the Arthur A. Dugoni School of Dentistry at Scholarly Commons. It has been accepted for inclusion in Dugoni School of Dentistry Faculty Articles by an authorized administrator of Scholarly Commons. For more information, please contact [email protected]. Authors Shu Chen Hung, Pei Rong Huang, Cassio Luiz Coutinho Almeida-Da-Silva, Kalina R. Atanasova, Ozlem Yilmaz, and David M. Ojcius This article is available at Scholarly Commons: https://scholarlycommons.pacific.edu/dugoni-facarticles/705 Version of Record: https://www.sciencedirect.com/science/article/pii/S1286457917301582 Manuscript_dd7f93413c97aff4865d54242a8b21e7 1 NLRX1 modulates differentially NLRP3 inflammasome activation 2 and NF-κB signaling during Fusobacterium nucleatum infection 3 4 5 Shu-Chen Hung 1, *, Pei-Rong Huang 2, Cássio Luiz Coutinho Almeida-da-Silva 1,3 , 6 Kalina R.
    [Show full text]
  • 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]
  • 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.
    [Show full text]
  • Coincidental Loss of DOCK8 Function in NLRP10-Deficient and C3H/Hej Mice Results in Defective Dendritic Cell Migration
    Coincidental loss of DOCK8 function in NLRP10-deficient and C3H/HeJ mice results in defective dendritic cell migration Jayendra Kumar Krishnaswamya,b,1, Arpita Singha,b,1, Uthaman Gowthamana,b, Renee Wua,b, Pavane Gorrepatia,b, Manuela Sales Nascimentoa,b, Antonia Gallmana,b, Dong Liua,b, Anne Marie Rhebergenb, Samuele Calabroa,b, Lan Xua,b, Patricia Ranneya,b, Anuj Srivastavac, Matthew Ransond, James D. Gorhamd, Zachary McCawe, Steven R. Kleebergere, Leonhard X. Heinzf, André C. Müllerf, Keiryn L. Bennettf, Giulio Superti-Furgaf, Jorge Henao-Mejiag, Fayyaz S. Sutterwalah, Adam Williamsi, Richard A. Flavellb,j,2, and Stephanie C. Eisenbartha,b,2 Departments of aLaboratory Medicine and bImmunobiology and jHoward Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520; cComputational Sciences, The Jackson Laboratory, Bar Harbor, ME 04609; dDepartment of Pathology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755; eNational Institute of Environmental Health Sciences, Research Triangle Park, NC 27709; fCeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; gInstitute for Immunology, Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; hInflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA 52241; and iThe Jackson Laboratory for Genomic Medicine, Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032 Contributed by Richard A. Flavell, January 28, 2015 (sent for review December 23, 2014; reviewed by Matthew L. Albert and Thirumala-Devi Kanneganti) Dendritic cells (DCs) are the primary leukocytes responsible for NLRP10 is the only NOD-like receptor (NLR) without a leu- priming T cells.
    [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]
  • A Role for the Nlr Family Members Nlrc4 and Nlrp3 in Astrocytic Inflammasome Activation and Astrogliosis
    A ROLE FOR THE NLR FAMILY MEMBERS NLRC4 AND NLRP3 IN ASTROCYTIC INFLAMMASOME ACTIVATION AND ASTROGLIOSIS Leslie C. Freeman A dissertation submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Curriculum of Genetics and Molecular Biology. Chapel Hill 2016 Approved by: Jenny P. Y. Ting Glenn K. Matsushima Beverly H. Koller Silva S. Markovic-Plese Pauline. Kay Lund ©2016 Leslie C. Freeman ALL RIGHTS RESERVED ii ABSTRACT Leslie C. Freeman: A Role for the NLR Family Members NLRC4 and NLRP3 in Astrocytic Inflammasome Activation and Astrogliosis (Under the direction of Jenny P.Y. Ting) The inflammasome is implicated in many inflammatory diseases but has been primarily studied in the macrophage-myeloid lineage. Here we demonstrate a physiologic role for nucleotide-binding domain, leucine-rich repeat, CARD domain containing 4 (NLRC4) in brain astrocytes. NLRC4 has been primarily studied in the context of gram-negative bacteria, where it is required for the maturation of pro-caspase-1 to active caspase-1. We show the heightened expression of NLRC4 protein in astrocytes in a cuprizone model of neuroinflammation and demyelination as well as human multiple sclerotic brains. Similar to macrophages, NLRC4 in astrocytes is required for inflammasome activation by its known agonist, flagellin. However, NLRC4 in astrocytes also mediate inflammasome activation in response to lysophosphatidylcholine (LPC), an inflammatory molecule associated with neurologic disorders. In addition to NLRC4, astrocytic NLRP3 is required for inflammasome activation by LPC. Two biochemical assays show the interaction of NLRC4 with NLRP3, suggesting the possibility of a NLRC4-NLRP3 co-inflammasome.
    [Show full text]
  • AIM2 and NLRC4 Inflammasomes Contribute with ASC to Acute Brain Injury Independently of NLRP3
    AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3 Adam Denesa,b,1, Graham Couttsb, Nikolett Lénárta, Sheena M. Cruickshankb, Pablo Pelegrinb,c, Joanne Skinnerb, Nancy Rothwellb, Stuart M. Allanb, and David Broughb,1 aLaboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Budapest, 1083, Hungary; bFaculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; and cInflammation and Experimental Surgery Unit, CIBERehd (Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas), Murcia Biohealth Research Institute–Arrixaca, University Hospital Virgen de la Arrixaca, 30120 Murcia, Spain Edited by Vishva M. Dixit, Genentech, San Francisco, CA, and approved February 19, 2015 (received for review November 18, 2014) Inflammation that contributes to acute cerebrovascular disease is or DAMPs, it recruits ASC, which in turn recruits caspase-1, driven by the proinflammatory cytokine interleukin-1 and is known causing its activation. Caspase-1 then processes pro–IL-1β to a to exacerbate resulting injury. The activity of interleukin-1 is regu- mature form that is rapidly secreted from the cell (5). The ac- lated by multimolecular protein complexes called inflammasomes. tivation of caspase-1 can also cause cell death (6). There are multiple potential inflammasomes activated in diverse A number of inflammasome-forming PRRs have been iden- diseases, yet the nature of the inflammasomes involved in brain tified, including NLR family, pyrin domain containing 1 (NLRP1); injury is currently unknown. Here, using a rodent model of stroke, NLRP3; NLRP6; NLRP7; NLRP12; NLR family, CARD domain we show that the NLRC4 (NLR family, CARD domain containing 4) containing 4 (NLRC4); AIM 2 (absent in melanoma 2); IFI16; and AIM2 (absent in melanoma 2) inflammasomes contribute to and RIG-I (5).
    [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]
  • Criteria for CAPS, Is It All in the Name? Markers, Urticaria-Like Rash and Arthralgia) Would Perform Well with These Four FCAS, but Only FCAS1 Is a CAPS
    ARD Online First, published on November 16, 2016 as 10.1136/annrheumdis-2016-210681 Ann Rheum Dis: first published as 10.1136/annrheumdis-2016-210681 on 16 November 2016. Downloaded from Correspondence Criteria for CAPS, is it all in the name? markers, urticaria-like rash and arthralgia) would perform well with these four FCAS, but only FCAS1 is a CAPS. The authors ‘ This paper1 describes an original work conducted by an fairly admitted that the number of CAPS cases and controls was International team of 16 recognised clinical experts in the field limited and not all possible differential diagnoses of CAPS may of autoinflammatory diseases. The aim of this consortium was have been included, potentially leading to an overestimation of fi ’ to develop diagnostic criteria for cryopyrin-associated periodic the speci city of the proposed model . fl syndrome (CAPS). They resulted in a model that indisputably is The issue of the disease name, re ecting either the main relevant to describe these rare and heterogeneous diseases symptoms or the molecular mechanisms of the condition, has fl among other autoinflammatory diseases. Their proposed CAPS been raised many times in the autoin ammatory diseases com- fi diagnosis criteria are primarily clinical. munity, and propositions for re ned taxonomy will shortly We would like to comment on the pathophysiological mech- emerge. The debate is not just semantic as differential thera- anism underlying ‘CAPS’. The NLRP3 gene encodes cryopyrin, peutic approaches can be taken according to the molecular the historical name of the NLRP3 protein, a key component of defect in cause in the condition presented by the patient.
    [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]
  • A Small-Molecule Inhibitor of the NLRP3 Inflammasome for the Treatment of Inflammatory Diseases
    ARTICLES A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases Rebecca C Coll1,2, Avril A B Robertson2, Jae Jin Chae3, Sarah C Higgins1, Raúl Muñoz-Planillo4, Marco C Inserra2,5, Irina Vetter2,5, Lara S Dungan1, Brian G Monks6, Andrea Stutz6, Daniel E Croker2, Mark S Butler2, Moritz Haneklaus1, Caroline E Sutton1, Gabriel Núñez4, Eicke Latz6–8, Daniel L Kastner3, Kingston H G Mills1, Seth L Masters9, Kate Schroder2, Matthew A Cooper2 & Luke A J O’Neill1 The NOD-like receptor (NLR) family, pyrin domain–containing protein 3 (NLRP3) inflammasome is a component of the inflammatory process, and its aberrant activation is pathogenic in inherited disorders such as cryopyrin-associated periodic syndrome (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer’s disease and atherosclerosis. We describe the development of MCC950, a potent, selective, small-molecule inhibitor of NLRP3. MCC950 blocked canonical and noncanonical NLRP3 activation at nanomolar concentrations. MCC950 specifically inhibited activation of NLRP3 but not the AIM2, NLRC4 or NLRP1 inflammasomes. MCC950 reduced interleukin-1b (IL-1b) production in vivo and attenuated the severity of experimental autoimmune encephalomyelitis (EAE), a disease model of multiple sclerosis. Furthermore, MCC950 treatment rescued neonatal lethality in a mouse model of CAPS and was active in ex vivo samples from individuals with Muckle–Wells syndrome. MCC950 is thus a potential therapeutic for NLRP3-associated syndromes, including autoinflammatory and autoimmune diseases, and a tool for further study of the NLRP3 inflammasome in human health and disease. The NLR family protein NLRP3 is an intracellular signaling molecule A role for NLRP3 in diseases of the central nervous system is that senses many pathogen-derived, environmental and host-derived emerging, and lung diseases have also been shown to be influenced factors1.
    [Show full text]