143 research outputs found

    Mechanisms of Gasdermin Family Members in Inflammasome Signaling and Cell Death

    Get PDF
    The Gasdermin (GSDM) family consists of Gasdermin A (GSDMA), Gasdermin B (GSDMB), Gasdermin C (GSDMC), Gasdermin D (GSDMD), Gasdermin E (GSDME) and Pejvakin (PJVK). GSDMD is activated by inflammasome-associated inflammatory caspases. Cleavage of GSDMD by human or mouse caspase-1, human caspase-4, human caspase-5, and mouse caspase-11 liberates the N-terminal effector domain from the C-terminal inhibitory domain. The N-terminal domain oligomerizes in the cell membrane and forms a pore of 10-16 nm in diameter, through which substrates of a smaller diameter, such as interleukin-1β and interleukin-18, are secreted. The increasing abundance of membrane pores ultimately leads to membrane rupture and pyroptosis, releasing the entire cellular content. Other than GSDMD, the N-terminal domain of all GSDMs, with the exception of PJVK, have the ability to form pores. There is evidence to suggest that GSDMB and GSDME are cleaved by apoptotic caspases. Here, we review the mechanistic functions of GSDM proteins with respect to their expression and signaling profile in the cell, with more focused discussions on inflammasome activation and cell death.S.F. was supported by a scholarship from the China Scholarship Council. S.M.M. is supported by the Australian National University, The Gretel and Gordon Bootes Medical Research Foundation and the National Health and Medical Research Council of Australia under Project grants (APP1141504 and APP1146864) and the R.G. Menzies Early Career Fellowship (APP1091544)

    Cathepsin B modulates lysosomal biogenesis and host defense against Francisella novicida infection

    Get PDF
    Lysosomal cathepsins regulate an exquisite range of biological functions, and their deregulation is associated with inflammatory, metabolic, and degenerative diseases in humans. In this study, we identified a key cell-intrinsic role for cathepsin B as a negative feedback regulator of lysosomal biogenesis and autophagy. Mice and macrophages lacking cathepsin B activity had increased resistance to the cytosolic bacterial pathogen Francisella novicida. Genetic deletion or pharmacological inhibition of cathepsin B down-regulated mechanistic target of rapamycin activity and prevented cleavage of the lysosomal calcium channel TRP ML1. These events drove transcription of lysosomal and autophagy genes via transcription factor EB, which increased lysosomal biogenesis and activation of autophagy initiation kinase ULK1 for clearance of the bacteria. Our results identified a fundamental biological function of cathepsin B in providing a checkpoint for homeostatic maintenance of lysosome populations and basic recycling functions in the cell

    Molecular mechanisms activating the NAIP‐NLRC4 inflammasome: Implications in infectious disease, autoinflammation, and cancer

    Get PDF
    Cytosolic innate immune sensing is a cornerstone of innate immunity in mammalian cells and provides a surveillance system for invading pathogens and endogenous danger signals. The NAIP‐NLRC4 inflammasome responds to cytosolic flagellin, and the inner rod and needle proteins of the type 3 secretion system of bacteria. This complex induces caspase‐1‐dependent proteolytic cleavage of the proinflammatory cytokines IL‐1β and IL‐18, and the pore‐forming protein gasdermin D, leading to inflammation and pyroptosis, respectively. Localized responses triggered by the NAIP‐NLRC4 inflammasome are largely protective against bacterial pathogens, owing to several mechanisms, including the release of inflammatory mediators, liberation of concealed intracellular pathogens for killing by other immune mechanisms, activation of apoptotic caspases, caspase‐7, and caspase‐8, and expulsion of an entire infected cell from the mammalian host. In contrast, aberrant activation of the NAIP‐NLRC4 inflammasome caused by de novo gain‐of‐function mutations in the gene encoding NLRC4 can lead to macrophage activation syndrome, neonatal enterocolitis, fetal thrombotic vasculopathy, familial cold autoinflammatory syndrome, and even death. Some of these clinical manifestations could be treated by therapeutics targeting inflammasome‐associated cytokines. In addition, the NAIP‐NLRC4 inflammasome has been implicated in the pathogenesis of colorectal cancer, melanoma, glioma, and breast cancer. However, no consensus has been reached on its function in the development of any cancer types. In this review, we highlight the latest advances in the activation mechanisms and structural assembly of the NAIP‐NLRC4 inflammasome, and the functions of this inflammasome in different cell types. We also describe progress toward understanding the role of the NAIP‐NLRC4 inflammasome in infectious diseases, autoinflammatory diseases, and cancer.Australian National University; National Health and Medical Research Council, Grant/Award Number: APP1141504,, APP1146864, APP1162103 and APP1163358; R.D. Wright Career Development Fellowship, Grant/Award Number: APP116202

    Bacillus cereus: Epidemiology, Virulence Factors, and Host-Pathogen Interactions

    Get PDF
    Bacillus cereus is an important human pathogen, and new findings have expanded our understanding of how this bacterium causes disease. B. cereus Hemolysin BL (HBL) and nonhemolytic enterotoxin (NHE) induce membrane pore formation, leading to activation of the NLRP3 inflammasome, systemic inflammation, and death. Lipopolysaccharide-induced tumor necrosis factor (TNF)-Îą factor (LITAF) and cell death-inducing P53 target 1 (CDIP1) are bona fide mammalian surface receptors of HBL. These newly identified toxin receptors and the NLRP3 inflammasome represent unique targets for potential future therapies against severe B. cereus infections. The toxin-producing bacterium Bacillus cereus is an important and neglected human pathogen and a common cause of food poisoning. Several toxins have been implicated in disease, including the pore-forming toxins hemolysin BL (HBL) and nonhemolytic enterotoxin (NHE). Recent work revealed that HBL binds to the mammalian surface receptors LITAF and CDIP1 and that both HBL and NHE induce potassium efflux and activate the NLRP3 inflammasome, leading to pyroptosis. These mammalian receptors, in part, contribute to inflammation and pathology. Other putative virulence factors of B. cereus include cytotoxin K, cereulide, metalloproteases, sphingomyelinase, and phospholipases. In this review, we highlight the latest progress in our understanding of B. cereus biology, epidemiology, and pathogenesis, and discuss potential new directions for research in this field.S.M.M. is supported by the Australian National University and the National Health and Medical Research Council of Australia under Project Grants (APP1141504, APP1146864, APP1162103 and APP1163358) and the R.D. Wright Career Development Fellowship (APP1162025). D.E.T. and S.M.M. are supported by Therapeutic Innovation Australia. D.E.T and A.M. are supported by The Gretel and Gordon Bootes Medical Research Foundation. A.M. is supported by a John Curtin School of Medical Research International PhD scholarshi

    Interferon-inducible guanylate-binding proteins at the interface of cell-autonomous immunity and inflammasome activation

    Get PDF
    Guanylate-binding proteins (GBPs) are essential components of cell-autonomous immunity. In response to IFN signaling, GBPs are expressed in the cytoplasm of immune and nonimmune cells, where they unleash their antimicrobial activity toward intracellular bacteria, viruses, and parasites. Recent studies have revealed that GBPs are essential for mediating activation of the caspase-1 inflammasome in response to the gram-negative bacteria Salmonella enterica serovar Typhimurium, Francisella novicida, Chlamydia muridarum, Chlamydia trachomatis, Legionella pneumophila, Vibrio cholerae, Enterobacter cloacae, and Citrobacter koseri. During infection with vacuolar-restricted gram-negative bacteria, GBPs disrupt the vacuolar membrane to ensure liberation of LPS for cytoplasmic detection by caspase-11 and the noncanonical NLRP3 inflammasome. In response to certain cytosolic bacteria, GBPs liberate microbial DNA for activation of the DNA-sensing AIM2 inflammasome. GBPs also promote the recruitment of antimicrobial proteins, including NADPH oxidase subunits and autophagy-associated proteins to the Mycobacteriumcontaining vacuole to mediate intracellular bacterial killing. Here, we provide an overview on the emerging relationship between GBPs and activation of the inflammasome in innate immunity to microbial pathogens.This work was supported by the U.S. National Institutes of Health (NIH) National Institute of Allergy and Infectious Diseases Grants AI101935, AI124346; NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases Grant AR056296; and NIH National Cancer Institute Grant CA163507 (to T.D.K.); the American Lebanese Syrian Associated Charities (to T.D.K.); and the R.G. Menzies Early Career Fellowship from the National Health and Medical Research Council of Australia (to S.M.M.

    Diverse and strain-specific metabolites patterns induced by fungal endophytes in grape cells of different varieties

    Get PDF
    The potential for endophytes to initiate changes in host secondary metabolism has been well documented. However, the mechanisms underlying endophyte-plant metabolic interactions are still poorly understood. Here, we analysed the effects of fungal endophytes on the metabolite profiles of grape cells from two cultivars: 'Cabernet Sauvignon' (CS) and 'Rose honey' (RH). Our results clearly showed that co-culture with endophytic fungi greatly modified the metabolic profiles in grape cells of both varieties. Treatments with endophytic fungal strains caused the numbers of detected metabolites to vary from 10 to 19 in CS cells and from 8 to 14 in RH cells. In addition, 5 metabolites were detected in all CS cell samples, while 4 metabolites were detected in all RH cell samples. Some endophytic fungal strains could even introduce novel metabolites into the co-cultured grape cells. The metabolic profiles of grape leaves shaped by endophytic fungi exhibited host selectivity and fungal strain specificity. In this assay, the fungal strains RH32 (Alternaria sp.) and MDR36 (Colletotrichum sp.) triggered an increased response of the detected metabolites, including the greatest increase in the metabolite contents in grape cells of both cultivars. No obvious effects in terms of metabolite numbers and contents in grape cells when co-cultured with fungal strains RH7 (Epicoccum sp.) and RH48 (Colletotrichum sp.) were observed. The results of this experiment suggest that endophytic fungi could be used to control the metabolic profiles of grapes and thus increase grape quality

    IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases

    Get PDF
    Inflammation underpins and contributes to the pathogenesis of many retinal degenerative diseases. The recruitment and activation of both resident microglia and recruited macrophages, as well as the production of cytokines, are key contributing factors for progressive cell death in these diseases. In particular, the interleukin 1 (IL-1) family consisting of both pro- and anti-inflammatory cytokines has been shown to be pivotal in the mediation of innate immunity and contribute directly to a number of retinal degenerations, including Age-Related Macular Degeneration (AMD), diabetic retinopathy, retinitis pigmentosa, glaucoma, and retinopathy of prematurity (ROP). In this review, we will discuss the role of IL-1 family members and inflammasome signaling in retinal degenerative diseases, piecing together their contribution to retinal disease pathology, and identifying areas of research expansion required to further elucidate their function in the retina

    Inflammasome activation causes dual recruitment of NLRC4 and NLRP3 to the same macromolecular complex.

    Get PDF
    Pathogen recognition by nucleotide-binding oligomerization domain-like receptor (NLR) results in the formation of a macromolecular protein complex (inflammasome) that drives protective inflammatory responses in the host. It is thought that the number of inflammasome complexes forming in a cell is determined by the number of NLRs being activated, with each NLR initiating its own inflammasome assembly independent of one another; however, we show here that the important foodborne pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) simultaneously activates at least two NLRs, whereas only a single inflammasome complex is formed in a macrophage. Both nucleotide-binding domain and leucine-rich repeat caspase recruitment domain 4 and nucleotide-binding domain and leucine-rich repeat pyrin domain 3 are simultaneously present in the same inflammasome, where both NLRs are required to drive IL-1β processing within the Salmonella-infected cell and to regulate the bacterial burden in mice. Superresolution imaging of Salmonella-infected macrophages revealed a macromolecular complex with an outer ring of apoptosis-associated speck-like protein containing a caspase activation and recruitment domain and an inner ring of NLRs, with active caspase effectors containing the pro-IL-1β substrate localized internal to the ring structure. Our data reveal the spatial localization of different components of the inflammasome and how different members of the NLR family cooperate to drive robust IL-1β processing during Salmonella infection.S.M.M was supported by a Cambridge International Scholarship. T.P.M was supported by a Wellcome Trust Research Career Development Fellowship (WT085090MA). This study was supported by Biotechnology and Biological Sciences Research Council (BBSRC) grants (BB/H003916/1 and BB/K006436/1) and a BBSRC Research Development Fellowship (BB/H021930/1) awarded to C.E.B.http://www.pnas.org/content/early/2014/05/05/1402911111.abstrac

    Actin polymerization as a key innate immune effector mechanism to control Salmonella infection.

    Get PDF
    Salmonellosis is one of the leading causes of food poisoning worldwide. Controlling bacterial burden is essential to surviving infection. Nucleotide-binding oligomerization domain-like receptors (NLRs), such as NLRC4, induce inflammasome effector functions and play a crucial role in controlling Salmonella infection. Inflammasome-dependent production of IL-1β recruits additional immune cells to the site of infection, whereas inflammasome-mediated pyroptosis of macrophages releases bacteria for uptake by neutrophils. Neither of these functions is known to directly kill intracellular salmonellae within macrophages. The mechanism, therefore, governing how inflammasomes mediate intracellular bacterial-killing and clearance in host macrophages remains unknown. Here, we show that actin polymerization is required for NLRC4-dependent regulation of intracellular bacterial burden, inflammasome assembly, pyroptosis, and IL-1β production. NLRC4-induced changes in actin polymerization are physically manifested as increased cellular stiffness, and leads to reduced bacterial uptake, production of antimicrobial molecules, and arrested cellular migration. These processes act in concert to limit bacterial replication in the cell and dissemination in tissues. We show, therefore, a functional link between innate immunity and actin turnover in macrophages that underpins a key host defense mechanism for the control of salmonellosis.Financial support for this work was provided by a Cambridge International Scholarship (to S.M.M.), European Research Council Starting Investigator Grant “LightTouch” 282060 (to J.R.G.), Biotechnology and Biological Sciences Research Council (BBSRC) Grants BB/H003916/1 and BB/K006436/1 and BBSRC Research Development Fellowship BB/ H021930/1 (to C.E.B.)This is the accepted manuscript. The final version is available from PNAS at http://www.pnas.org/content/111/49/17588.abstract

    Bacillus cereus non-haemolytic enterotoxin activates the NLRP3 inflammasome

    Get PDF
    Inflammasomes are important for host defence against pathogens and homeostasis with commensal microbes. Here, we show non-haemolytic enterotoxin (NHE) from the neglected human foodborne pathogen Bacillus cereus is an activator of the NLRP3 inflammasome and pyroptosis. NHE is a non-redundant toxin to haemolysin BL (HBL) despite having a similar mechanism of action. Via a putative transmembrane region, subunit C of NHE initiates binding to the plasma membrane, leading to the recruitment of subunit B and subunit A, thus forming a tripartite lytic pore that is permissive to efflux of potassium. NHE mediates killing of cells from multiple lineages and hosts, highlighting a versatile functional repertoire in different host species. These data indicate that NHE and HBL operate synergistically to induce inflammation and show that multiple virulence factors from the same pathogen with conserved function and mechanism of action can be exploited for sensing by a single inflammasome
    • …
    corecore