Vaccinia Virus F1L Protein Promotes Virulence by Inhibiting Inflammasome Activation

Total Page:16

File Type:pdf, Size:1020Kb

Vaccinia Virus F1L Protein Promotes Virulence by Inhibiting Inflammasome Activation Vaccinia virus F1L protein promotes virulence by inhibiting inflammasome activation Motti Gerlica,1, Benjamin Faustina,1, Antonio Postigob, Eric Chi-Wang Yua, Martina Proella,c, Naran Gombosurena, Maryla Krajewskaa, Rachel Flynnd, Michael Croftd, Michael Wayb, Arnold Satterthwaita, Robert C. Liddingtona, Shahram Salek-Ardakanid,2, Shu-ichi Matsuzawaa, and John C. Reeda,3 aSanford-Burnham Medical Research Institute, La Jolla, CA 92037; bCancer Research UK London Research Institute, London WC2A 3LY, United Kingdom; cDepartment of Molecular Biology, University of Salzburg, 5020 Salzburg, Austria; and dDivision of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037 Edited by Elliott Kieff, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, and approved March 22, 2013 (received for review October 8, 2012) Host innate immune responses to DNA viruses involve members of Bcl-2 family members, several of these viral homologs were dis- the nucleotide-binding domain, leucine-rich repeat and pyrin do- covered to adopt an α-helical fold similar to Bcl-2 (13-17). However, main containing protein (NLRP) family, which form “inflamma- not all of the VACV-encoded Bcl-2–like proteins possess anti- somes” that activate caspase-1, resulting in proteolytic activation apoptotic activity (18), implying that they have other functions. of cytokines interleukin (IL)-1β and IL-18. We hypothesized that Indeed, the viral Bcl-2–like proteins A52, B14, N1, and K7 were DNA viruses would target inflammasomes to overcome host de- found to inhibit NF-κB pathways at various checkpoints (11). fense. A Vaccinia virus (VACV) B-cell CLL/lymphoma 2 (Bcl-2) homo- Recently, cellular Bcl-2 family proteins have been demonstrated log, F1L, was demonstrated to bind and inhibit the NLR family to interact with NLRs to impact innate immunity responses member NLRP1 in vitro. Moreover, infection of macrophages in cul- (19, 20). We therefore hypothesized that viral Bcl-2 homologs ture with virus lacking F1L (ΔF1L) caused increased caspase-1 activa- encoded by VACV might similarly inhibit inflammasome signaling tion and IL-1β secretion compared with wild-type virus. Virulence of during infection. Our data provide evidence that the viral Bcl-2 ΔF1L virus was attenuated in vivo, causing altered febrile responses, homolog F1L (but not N1L) is a suppressor of NLR family proteins increased proteolytic processing of caspase-1, and more rapid in- involved in IL-1β activation. Furthermore, by testing genetically flammation in lungs of infected mice without affecting cell death engineered VACV in a mouse model, our findings also demon- or virus replication. Furthermore, we found that a hexapeptide from strate a role for NLR suppression in the pathogenic mechanism by F1L is necessary and sufficient for inhibiting the NLRP1 inflamma- which F1L contributes to virulence in vivo. some in vitro, thus identifying a peptidyl motif required for binding and inhibiting NLRP1. The functional importance of this NLRP1-bind- Results ing motif was further confirmed by studies of recombinant ΔF1L VACV Bcl-2 Homolog, F1L, Binds NLRP1. Of the viral Bcl-2 homologs viruses reconstituted either with the wild-type F1L or a F1L mutant encoded by VACV, only F1L and N1L have been reported to that fails to bind NLRP1. Cellular infection with wild-type F1L recon- display antiapoptotic activity and to bind proapoptotic members of stituted virus-suppressed IL-1β production, whereas mutant F1L did the cellular Bcl-2 family such as Bcl-2–associated X protein (Bax) not. In contrast, both wild-type and mutant versions of F1L equally (14, 18, 21). We explored the ability of F1L to bind various NLR suppressed apoptosis. In vivo, the NLR nonbinding F1L mutant virus family members, making comparisons with apoptosis-associated exhibited an attenuated phenotype similar to ΔF1L virus, thus con- speck-like protein containing a caspase recruitment domain (ASC), firming the importance of F1L interactions with NLRP1 for viral path- absent in melanoma 2 (AIM2), and other proteins involved in in- ogenicity in mice. Altogether, these findings reveal a unique viral nate immune defense against viruses. In cell transfection experi- mechanism for evading host innate immune responses. ments, F1L coimmunoprecipitated with NLRP1, a NLR member inhibited by Bcl-2 and Bcl-XL (20), but not with NLR family virus infection | virology | innate immunity | poxviruses members NLRP3 and nucleotide-binding oligomerization domain- containing protein 2 (NOD2) (NLRC2) and not with AIM2 or ASC (Fig. 1A and Fig. S1A). Colocalization of F1L with NLRP1 ucleotide-binding domain and leucine-rich repeat containing was also demonstrated by immunofluorescence confocal micros- Nreceptors (NLRs) constitute a large family of intracellular copy (Fig. S1C). In contrast to F1L, the viral N1L protein did not innate immunity proteins involved in host defense (1). Upon ac- associate with NLRP1 as determined by coimmunoprecipitation tivation, NLRs form large protein complexes called “inflamma- fl ” (co-IP) assay and by immuno uorescence confocal microscopy somes that bind and activate caspase-1 family proteases, resulting (Fig. 1A and Fig. S1C). in proteolytic activation of proinflammatory cytokines (which in- fi – β – The interaction of F1L with NLRP1 was further con rmed by clude pro IL-1 and pro IL-18) and sometimes causing a caspase- in vitro protein binding assays, using GST-tagged recombinant 1–dependent form of cell death known as pyroptosis (2, 3). Members of the NLR family possess a conserved architecture that includes leucine-rich repeats (LRRs) that are thought to act as Author contributions: M.G., B.F., M.W., S.M., and J.C.R. designed research; M.G., B.F., A.P., receptors for various pathogen-derived molecules. Certain NLRs, M.P., R.F., S.S.-A., and S.M. performed research; M.G., B.F., A.P., E.C.-W.Y., N.G., M.W., and several other types of innate immunity proteins, are involved A.S., and R.C.L. contributed new reagents/analytic tools; M.G., B.F., A.P., M.P., M.K., M.C., in detecting foreign double-stranded DNA (dsDNA) in cells, a M.W., S.S.-A., and J.C.R. analyzed data; and M.G., B.F., and J.C.R. wrote the paper. function linked to host defense against DNA viruses (4–9). The authors declare no conflict of interest. Poxviruses constitute a viridae known for their large, complex This article is a PNAS Direct Submission. DNA genomes. Vaccinia virus (VACV) is a prototypical member of 1M.G. and B.F. contributed equally to this work. the poxvirus family, which replicates in the cytoplasm of host cells 2Present address: Department of Pathology, Immunology, and Laboratory Medicine, Uni- and encodes numerous proteins that manipulate the host response versity of Florida, Gainesville, FL 32610. to infection (10). VACV encodes several viral homologs of the 3To whom correspondence should be addressed. E-mail: reedoffice@sanfordburnham. cellular antiapoptotic protein B-cell CLL/lymphoma 2 (Bcl-2), org. including A46R, A52R, B14R, C1L, C6L, C16/B22R, F1L, K7R, This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. N1L, and N2L (11, 12). Despite low sequence similarity to cellular 1073/pnas.1215995110/-/DCSupplemental. 7808–7813 | PNAS | May 7, 2013 | vol. 110 | no. 19 www.pnas.org/cgi/doi/10.1073/pnas.1215995110 Downloaded by guest on September 28, 2021 A B reconstituted the NLRP1 inflammasome in vitro by using purified recombinant proteins, showing that the combination of NLRP1 ligand muramyl dipeptide (MDP) and ATP induces NLRP1 6 oligomerization and caspase-1 activation in a Bcl-2/Bcl-X - 5 L suppressible manner (22, 23). Adding recombinant F1L to these in 4 180 vitro reactions suppressed NLRP1-driven caspase-1 activity, whereas 3 N1L and GST control proteins did not (Fig. 1B). Comparison of 2 37 various fragments of F1L (truncated from the N terminus) sug- 1 (fold stimulation) gested that residues between aa 24 and 48 of F1L are required for 180 Caspase-1 activity 0 NLRP1 suppression (Fig. 1B), which correlated with binding to CNTL N1L GST (24-206) NLRP1 (Fig. S2A). Additional experiments using a GST-F1L F1L (1-206)L (48-206)L (74-206) 37 F1L F1 F1 F1L (90-206) (1–47) fusion protein showed that the first 47 amino acids of F1L are sufficient for suppressing NLRP1 activity in vitro (Fig. 1C). +MDP/ATP C 175 Unstimulated D 350 Next, we chemically synthesized peptides (Table S1) corre- 150 300 sponding to residues 1–47 of F1L and various shorter segments, 125 250 examining their effects on the reconstituted NLRP1 inflamma- 100 200 some in vitro. The addition of the F1L peptide 1–47 and shorter 75 150 peptides (residues 22–47 and 27–37) inhibited NLRP1-mediated (RFU/min) 50 (RFU/min) 100 caspase-1 activation to baseline levels (Fig. 1D and Fig. S2B). Caspase-1 activity 25 Caspase-1 activity 50 0 However, these F1L peptides were neither active against the 0 Δ N1L F1L N1L F1L gain-of-function mutant NLRP LRR (which does not bind F1L CNTLBcl-2 CNTLBcl-2 fi F1L(1-47)F1L(1-35) F1L(1-47)F1L(1-35) No peptide protein) nor against puri ed active recombinant caspase-1, F1Lp (27-37)F1Lp (32-37)F1Lp (22-35)F1Lp (22-34) showing NLRP1 dependence (Fig. S2 C and D). Furthermore, the F1L peptide (22–47) binds to NLRP1, as determined by EF120 350 fluorescence polarization assay using a fluorochrome-conjugated 100 300 E fl 80 250 peptide (Fig. S2 ). In addition to using a uorigenic peptide 200 substrate (Ac-WEHD-AMC) to monitor caspase-1 activity, we 60 150 also assessed NLRP1-induced proteolytic processing of caspase-1 40 (RFU/min) 100 ATP binding ATP (% inhibition) by immunoblotting, showing that both synthetic F1L peptide 20 50 – – Caspase-1 activity (22 47) and recombinant GST-F1L (1 47) protein inhibited 0 0 proteolytic processing of caspase-1 driven by NLRP1, but not by N1L F1L C7A GST L V33A 32-3717-3716-19 Δ F F1 F1L mutant constitutively active NLRP1 LRR (Fig.
Recommended publications
  • United States Patent (19) 11 Patent Number: 5,766,598 Paoletti Et Al
    USOO5766598A United States Patent (19) 11 Patent Number: 5,766,598 Paoletti et al. 45) Date of Patent: Jun. 16, 1998 54 RECOMBINANTATTENUATED ALVAC Murphy. F., 1996. "Virus Taxonomy", in Fields Virology, CANARYPOXVIRUS EXPRESSION Third Edition, Fields et al., eds. Lippincott-Raven Publish WECTORS CONTAINING HETEROLOGOUS ers. Philadelphia, pp. 15-57. DNA SEGMENTS ENCODNG LENTWRAL Taylor et al., 1991. Vaccine 9(3):190-193. GENE PRODUCTS Berman et al., 1990, Nature 345:622-625. Girard et al., 1991 Proc. Natl. Acad. Sci., USA 88:542-546. 75 Inventors: Enzo Paoletti, Delmar; James Tartaglia. Schenectady; William Irvin Hu et al., 1986. Nature 320:537-540. Cox. Troy, all of N.Y. Ho et al., 1990, Eur, J. Immunol. 20:477-483. Virology, vol. 173, No. 1, issued Nov. 1989, S. Dallo et al. 73) Assignee: Wirogenetics Corporation, Troy, N.Y. "Humoral Immune Response Elicited by Highly Attenuated Variants of Vaccinia Virus and by an Attenuated Recombi nant Expressing HIV-1 Envelope Protein". pp. 323-328 21 Appl. No.: 303,275 (entire document). (22 Filed: Sep. 7, 1994 Virology, vol. 152, issued 1986. M.E. Perkus et al., “Inser tion and Deletion Mutants of Vaccinia Virus". pp. 285-297 Related U.S. Application Data (entire document). Nature, vol. 317, issued 31 Oct. 1985, R.M.L. Buller et al., 63 Continuation of Ser. No. 897,382, Jun. 11, 1992, abandoned, which is a continuation-in-part of Ser. No. 715,921, Jun. 14, "Decreased Virulence of Recombinant Vaccinia Virus 1991, abandoned, and Ser. No. 847,951, Mar. 6, 1992, which Expression Vectors is Associated With a Thymidine is a continuation-in-part of Ser.
    [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]
  • 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]
  • Transcriptomic Profiles of High and Low Antibody Responders to Smallpox
    Genes and Immunity (2013) 14, 277–285 & 2013 Macmillan Publishers Limited All rights reserved 1466-4879/13 www.nature.com/gene ORIGINAL ARTICLE Transcriptomic profiles of high and low antibody responders to smallpox vaccine RB Kennedy1,2, AL Oberg1,3, IG Ovsyannikova1,2, IH Haralambieva1,2, D Grill1,3 and GA Poland1,2 Despite its eradication over 30 years ago, smallpox (as well as other orthopox viruses) remains a pathogen of interest both in terms of biodefense and for its use as a vector for vaccines and immunotherapies. Here we describe the application of mRNA-Seq transcriptome profiling to understanding immune responses in smallpox vaccine recipients. Contrary to other studies examining gene expression in virally infected cell lines, we utilized a mixed population of peripheral blood mononuclear cells in order to capture the essential intercellular interactions that occur in vivo, and would otherwise be lost, using single cell lines or isolated primary cell subsets. In this mixed cell population we were able to detect expression of all annotated vaccinia genes. On the host side, a number of genes encoding cytokines, chemokines, complement factors and intracellular signaling molecules were downregulated upon viral infection, whereas genes encoding histone proteins and the interferon response were upregulated. We also identified a small number of genes that exhibited significantly different expression profiles in subjects with robust humoral immunity compared with those with weaker humoral responses. Our results provide evidence that differential gene regulation patterns may be at work in individuals with robust humoral immunity compared with those with weaker humoral immune responses. Genes and Immunity (2013) 14, 277–285; doi:10.1038/gene.2013.14; published online 18 April 2013 Keywords: Next-generation sequencing; mRNA-Seq; vaccinia virus; smallpox vaccine INTRODUCTION these 44 subjects had two samples (uninfected and vaccinia Vaccinia virus (VACV) is the immunologically cross-protective infected).
    [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]
  • PF a Colonne V 5 2020 12 16.Xlsx
    Istanza per il riconoscimento dell'esenzione della Tassa Automobilistica Ex Art 7 Comma 4 della Legge Regionale 12 Maggio 2020 n. 9 Elenco delle richieste di rimborso Persone Fisiche Le istanze sono riportate nell'elenco ordinate in modo crescente. Il motivo dell'eventuale rifiuto è solo perché non è stato trovato un versamento corrispondente al veicolo indicato per l'annualità 2020.
    [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]