Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(4): eadh3409, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38277448

ABSTRACT

The innate immune response contributes to the development or attenuation of acute and chronic diseases, including cancer. Microbial DNA and mislocalized DNA from damaged host cells can activate different host responses that shape disease outcomes. Here, we show that mice and humans lacking a single allele of the DNA repair protein Ku70 had increased susceptibility to the development of intestinal cancer. Mechanistically, Ku70 translocates from the nucleus into the cytoplasm where it binds to cytosolic DNA and interacts with the GTPase Ras and the kinase Raf, forming a tripartite protein complex and docking at Rab5+Rab7+ early-late endosomes. This Ku70-Ras-Raf signalosome activates the MEK-ERK pathways, leading to impaired activation of cell cycle proteins Cdc25A and CDK1, reducing cell proliferation and tumorigenesis. We also identified the domains of Ku70, Ras, and Raf involved in activating the Ku70 signaling pathway. Therapeutics targeting components of the Ku70 signalosome could improve the treatment outcomes in cancer.


Subject(s)
Neoplasms , Signal Transduction , Animals , Humans , Mice , Cell Proliferation , DNA , MAP Kinase Signaling System , Neoplasms/genetics
2.
Trends Biochem Sci ; 48(10): 883-893, 2023 10.
Article in English | MEDLINE | ID: mdl-37567806

ABSTRACT

Guanylate-binding proteins (GBPs) are a family of intracellular proteins which have diverse biological functions, including pathogen sensing and host defense against infectious disease. These proteins are expressed in response to interferon (IFN) stimulation and can localize and target intracellular microbes (e.g., bacteria and viruses) by protein trafficking and membrane binding. These properties contribute to the ability of GBPs to induce inflammasome activation, inflammation, and cell death, and to directly disrupt pathogen membranes. Recent biochemical studies have revealed that human GBP1, GBP2, and GBP3 can directly bind to the lipopolysaccharide (LPS) of Gram-negative bacteria. In this review we discuss emerging data highlighting the functional versatility of GBPs, with a focus on their molecular mechanisms of pattern recognition and antimicrobial activity.


Subject(s)
Anti-Infective Agents , Carrier Proteins , Humans , GTP-Binding Proteins/chemistry , Inflammasomes/metabolism , Bacteria/metabolism , Anti-Infective Agents/pharmacology
3.
EMBO Rep ; 24(6): e54600, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37073791

ABSTRACT

Inflammasome signaling is a central pillar of innate immunity triggering inflammation and cell death in response to microbes and danger signals. Here, we show that two virulence factors from the human bacterial pathogen Clostridium perfringens are nonredundant activators of the NLRP3 inflammasome in mice and humans. C. perfringens lecithinase (also known as phospolipase C) and C. perfringens perfringolysin O induce distinct mechanisms of activation. Lecithinase enters LAMP1+ vesicular structures and induces lysosomal membrane destabilization. Furthermore, lecithinase induces the release of the inflammasome-dependent cytokines IL-1ß and IL-18, and the induction of cell death independently of the pore-forming proteins gasdermin D, MLKL and the cell death effector protein ninjurin-1 or NINJ1. We also show that lecithinase triggers inflammation via the NLRP3 inflammasome in vivo and that pharmacological blockade of NLRP3 using MCC950 partially prevents lecithinase-induced lethality. Together, these findings reveal that lecithinase activates an alternative pathway to induce inflammation during C. perfringens infection and that this mode of action can be similarly exploited for sensing by a single inflammasome.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Clostridium perfringens/metabolism , Virulence Factors , Inflammation , Interleukin-1beta/metabolism , Nerve Growth Factors , Cell Adhesion Molecules, Neuronal
4.
Life Sci Alliance ; 6(6)2023 06.
Article in English | MEDLINE | ID: mdl-36944419

ABSTRACT

Povidone-iodine (PVP-I) inactivates a broad range of pathogens. Despite its widespread use over decades, the safety of PVP-I remains controversial. Its extended use in the current SARS-CoV-2 virus pandemic urges the need to clarify safety features of PVP-I on a cellular level. Our investigation in epithelial, mesothelial, endothelial, and innate immune cells revealed that the toxicity of PVP-I is caused by diatomic iodine (I2), which is rapidly released from PVP-I to fuel organic halogenation with fast first-order kinetics. Eukaryotic toxicity manifests at below clinically used concentrations with a threshold of 0.1% PVP-I (wt/vol), equalling 1 mM of total available I2 Above this threshold, membrane disruption, loss of mitochondrial membrane potential, and abolition of oxidative phosphorylation induce a rapid form of cell death we propose to term iodoptosis. Furthermore, PVP-I attacks lipid rafts, leading to the failure of tight junctions and thereby compromising the barrier functions of surface-lining cells. Thus, the therapeutic window of PVP-I is considerably narrower than commonly believed. Our findings urge the reappraisal of PVP-I in clinical practice to avert unwarranted toxicity whilst safeguarding its benefits.


Subject(s)
Anti-Infective Agents, Local , COVID-19 , Iodine , Humans , Povidone-Iodine/pharmacology , Povidone-Iodine/therapeutic use , Anti-Infective Agents, Local/pharmacology , Anti-Infective Agents, Local/therapeutic use , Iodine/pharmacology , SARS-CoV-2 , Cell Death
5.
EMBO J ; 42(6): e112558, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36762431

ABSTRACT

Moraxella catarrhalis is an important human respiratory pathogen and a major causative agent of otitis media and chronic obstructive pulmonary disease. Toll-like receptors contribute to, but cannot fully account for, the complexity of the immune response seen in M. catarrhalis infection. Using primary mouse bone marrow-derived macrophages to examine the host response to M. catarrhalis infection, our global transcriptomic and targeted cytokine analyses revealed activation of immune signalling pathways by both membrane-bound and cytosolic pattern-recognition receptors. We show that M. catarrhalis and its outer membrane vesicles or lipooligosaccharide (LOS) can activate the cytosolic innate immune sensor caspase-4/11, gasdermin-D-dependent pyroptosis, and the NLRP3 inflammasome in human and mouse macrophages. This pathway is initiated by type I interferon signalling and guanylate-binding proteins (GBPs). We also show that inflammasomes and GBPs, particularly GBP2, are required for the host defence against M. catarrhalis in mice. Overall, our results reveal an essential role for the interferon-inflammasome axis in cytosolic recognition and immunity against M. catarrhalis, providing new molecular targets that may be used to mitigate pathological inflammation triggered by this pathogen.


Subject(s)
Caspases , Inflammasomes , Mice , Humans , Animals , Caspases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Moraxella catarrhalis/metabolism , Carrier Proteins , Immunity, Innate
6.
Nat Commun ; 13(1): 4395, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906252

ABSTRACT

Inflammasomes are cytosolic signaling complexes capable of sensing microbial ligands to trigger inflammation and cell death responses. Here, we show that guanylate-binding proteins (GBPs) mediate pathogen-selective inflammasome activation. We show that mouse GBP1 and GBP3 are specifically required for inflammasome activation during infection with the cytosolic bacterium Francisella novicida. We show that the selectivity of mouse GBP1 and GBP3 derives from a region within the N-terminal domain containing charged and hydrophobic amino acids, which binds to and facilitates direct killing of F. novicida and Neisseria meningitidis, but not other bacteria or mammalian cells. This pathogen-selective recognition by this region of mouse GBP1 and GBP3 leads to pathogen membrane rupture and release of intracellular content for inflammasome sensing. Our results imply that GBPs discriminate between pathogens, confer activation of innate immunity, and provide a host-inspired roadmap for the design of synthetic antimicrobial peptides that may be of use against emerging and re-emerging pathogens.


Subject(s)
Carrier Proteins , Inflammasomes , Animals , Bacteria/metabolism , Carrier Proteins/metabolism , Cytosol/metabolism , GTP-Binding Proteins/metabolism , Immunity, Innate , Inflammasomes/metabolism , Mammals/metabolism , Mice , Signal Transduction
7.
Sci Immunol ; 7(71): eabm1803, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35594341

ABSTRACT

Clostridium species are a group of Gram-positive bacteria that cause diseases in humans, such as food poisoning, botulism, and tetanus. Here, we analyzed 10 different Clostridium species and identified that Clostridium septicum, a pathogen that causes sepsis and gas gangrene, activates the mammalian cytosolic inflammasome complex in mice and humans. Mechanistically, we demonstrate that α-toxin secreted by C. septicum binds to glycosylphosphatidylinositol (GPI)-anchored proteins on the host plasma membrane, oligomerizing and forming a membrane pore that is permissive to efflux of magnesium and potassium ions. Efflux of these cytosolic ions triggers the activation of the innate immune sensor NLRP3, inducing activation of caspase-1 and gasdermin D, secretion of the proinflammatory cytokines interleukin-1ß and interleukin-18, pyroptosis, and plasma membrane rupture via ninjurin-1. Furthermore, α-toxin of C. septicum induces rapid inflammasome-mediated lethality in mice and pharmacological inhibition of the NLRP3 inflammasome using MCC950 prevents C. septicum-induced lethality. Overall, our results reveal that cytosolic innate sensing of α-toxin is central to the recognition of C. septicum infection and that therapeutic blockade of the inflammasome pathway may prevent sepsis and death caused by toxin-producing pathogens.


Subject(s)
Bacterial Toxins , GPI-Linked Proteins , Inflammasomes , Animals , Bacterial Toxins/metabolism , Clostridium septicum/chemistry , GPI-Linked Proteins/metabolism , Glycosylphosphatidylinositols/metabolism , Inflammasomes/metabolism , Mammals/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis
8.
Trends Cell Biol ; 31(11): 924-939, 2021 11.
Article in English | MEDLINE | ID: mdl-34284921

ABSTRACT

Organelles are critical structures in mediating the assembly and activation of inflammasomes in mammalian cells, resulting in inflammation and cell death. Assembly of inflammasomes can occur at the mitochondria, endoplasmic reticulum, nucleus, trans-Golgi network, or pathogen surface, facilitated by the overarching architecture of the cytoskeleton. NLRP3 and Pyrin inflammasome sensors may form smaller speckles and converge on a single larger speck at the microtubule-organizing center (MTOC). This signaling hub activates multiple mammalian inflammatory and apoptotic caspases, cytokine substrates, the pore-forming protein gasdermin D, and the plasma membrane rupture protein ninjurin-1 (NINJ1), allowing pyroptosis, cellular disintegration, and inflammation to ensue. In this review, we highlight the role of mammalian cell types and organellar architectures in executing inflammasome responses.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Caspases/metabolism , Cell Adhesion Molecules, Neuronal , Humans , Inflammasomes/metabolism , Inflammation , Mammals/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Growth Factors , Pyroptosis
9.
Nat Immunol ; 21(11): 1470, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32913347

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Nat Commun ; 11(1): 760, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029733

ABSTRACT

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.


Subject(s)
Bacillus cereus/pathogenicity , Enterotoxins/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Bacterial Proteins/toxicity , Cell Line , Enterotoxins/chemistry , Female , Hemolysin Proteins/toxicity , Host Microbial Interactions , Host Specificity , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroptosis/drug effects , Virulence Factors/toxicity
12.
Nat Microbiol ; 4(2): 362-374, 2019 02.
Article in English | MEDLINE | ID: mdl-30531979

ABSTRACT

Host recognition of microbial components is essential in mediating an effective immune response. Cytosolic bacteria must secure entry into the host cytoplasm to facilitate replication and, in doing so, liberate microbial ligands that activate cytosolic innate immune sensors and the inflammasome. Here, we identified a multicomponent enterotoxin, haemolysin BL (HBL), that engages activation of the inflammasome. This toxin is highly conserved among the human pathogen Bacillus cereus. The three subunits of HBL bind to the cell membrane in a linear order, forming a lytic pore and inducing activation of the NLRP3 inflammasome, secretion of interleukin-1ß and interleukin-18, and pyroptosis. Mechanistically, the HBL-induced pore results in the efflux of potassium and triggers the activation of the NLRP3 inflammasome. Furthermore, HBL-producing B. cereus induces rapid inflammasome-mediated mortality. Pharmacological inhibition of the NLRP3 inflammasome using MCC950 prevents B. cereus-induced lethality. Overall, our results reveal that cytosolic sensing of a toxin is central to the innate immune recognition of infection. Therapeutic modulation of this pathway enhances host protection against deadly bacterial infections.


Subject(s)
Bacillus cereus/immunology , Bacterial Proteins/immunology , Enterotoxins/immunology , Hemolysin Proteins/immunology , Inflammasomes/metabolism , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Cells, Cultured , Culture Media, Conditioned , Enterotoxins/chemistry , Enterotoxins/metabolism , Female , Hemolysin Proteins/metabolism , Immunity, Innate , Macrophages/immunology , Macrophages/pathology , Macrophages/ultrastructure , Male , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Protein Multimerization , Pyroptosis , Survival Analysis
13.
Front Neural Circuits ; 12: 99, 2018.
Article in English | MEDLINE | ID: mdl-30483067

ABSTRACT

The mammalian basal forebrain (BF), a heterogenous structure providing the primary cholinergic inputs to cortical and limbic structures, plays a crucial role in various physiological processes such as learning/memory and attention. Despite the involvement of the BF cholinergic neurons (BFCNs) in olfaction related memory has been reported, the underlying neural circuits remain poorly understood. Here, we combined viral trans-synaptic tracing systems and ChAT-cre transgenic mice to systematically reveal the relationship between the olfactory system and the different subsets of BFCNs. The retrograde adeno-associated virus and rabies virus (AAV-RV) tracing showed that different subregional BFCNs received diverse inputs from multiple olfactory cortices. The cholinergic neurons in medial and caudal horizontal diagonal band Broca (HDB), magnocellular preoptic area (MCPO) and ventral substantia innominate (SI; hereafter HMS complex, HMSc) received the inputs from the entire olfactory system such as the olfactory bulb (OB), anterior olfactory nucleus (AON), entorhinal cortex (ENT), basolateral amygdala and especially the piriform cortex (PC) and hippocampus (HIP); while medial septum (MS/DB) and a part of rostral HDB (hereafter MS/DB complex, MS/DBc), predominantly from HIP; and nucleus basalis Meynert (NBM) and dorsal SI (hereafter NBM complex, NBMc), mainly from the central amygdala. The anterograde vesicular stomatitis virus (VSV) tracing further validated that the major target of the OB to the BF is HMSc. To correlate these structural relations between the BFCNs and olfactory functions, the neurons activated in the BF during olfaction related task were mapped with c-fos immunostaining. It was found that some of the BFCNs were activated in go/no-go olfactory discrimination task, but with different activated patterns. Interestingly, the BFCNs in HMSc were more significantly activated than the other subregions. Therefore, our data have demonstrated that among the different subgroups of BFCNs, HMSc is more closely related to the olfactory system, both structurally and functionally. This work provides the evidence for distinct roles of different subsets of BFNCs in olfaction associated memory.


Subject(s)
Basal Forebrain/cytology , Basal Forebrain/physiology , Cholinergic Neurons/physiology , Memory/physiology , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Animals , Basal Forebrain/chemistry , Cholinergic Neurons/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Bulb/chemistry , Smell/physiology
14.
J Mol Biol ; 430(18 Pt B): 3068-3080, 2018 09 14.
Article in English | MEDLINE | ID: mdl-29990470

ABSTRACT

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-1ß 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.


Subject(s)
Inflammasomes/metabolism , Multigene Family , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Biomarkers , Cell Death/genetics , Gene Expression Regulation , Humans , Immune System/immunology , Immune System/metabolism , Neoplasm Proteins/chemistry , Receptors, Pattern Recognition/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...