Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Science ; 384(6693): eadl2016, 2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38635718

RÉSUMÉ

Infectious diseases continue to claim many lives. Prevention of morbidity and mortality from these diseases would benefit not just from new medicines and vaccines but also from a better understanding of what constitutes protective immunity. Among the major immune signals that mobilize host defense against infection is interferon-γ (IFN-γ), a protein secreted by lymphocytes. Forty years ago, IFN-γ was identified as a macrophage-activating factor, and, in recent years, there has been a resurgent interest in IFN-γ biology and its role in human defense. Here we assess the current understanding of IFN-γ, revisit its designation as an "interferon," and weigh its prospects as a therapeutic against globally pervasive microbial pathogens.


Sujet(s)
Maladies transmissibles , Interféron gamma , Humains , Maladies transmissibles/immunologie , Maladies transmissibles/thérapie , Interféron gamma/génétique , Interféron gamma/métabolisme , Interféron gamma/usage thérapeutique , Immunité/génétique
2.
Science ; 383(6686): eabm9903, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38422126

RÉSUMÉ

All living organisms deploy cell-autonomous defenses to combat infection. In plants and animals, large supramolecular complexes often activate immune proteins for protection. In this work, we resolved the native structure of a massive host-defense complex that polymerizes 30,000 guanylate-binding proteins (GBPs) over the surface of gram-negative bacteria inside human cells. Construction of this giant nanomachine took several minutes and remained stable for hours, required guanosine triphosphate hydrolysis, and recruited four GBPs plus caspase-4 and Gasdermin D as a cytokine and cell death immune signaling platform. Cryo-electron tomography suggests that GBP1 can adopt an extended conformation for bacterial membrane insertion to establish this platform, triggering lipopolysaccharide release that activated coassembled caspase-4. Our "open conformer" model provides a dynamic view into how the human GBP1 defense complex mobilizes innate immunity to infection.


Sujet(s)
Bactéries , Infections bactériennes , Membrane cellulaire , Protéines G , , Humains , Cytokines/composition chimique , Tomographie en microscopie électronique , Protéines G/composition chimique , Guanosine triphosphate/composition chimique , Hydrolyse , Immunité cellulaire , Cryomicroscopie électronique , Gasdermines/composition chimique , Protéines de liaison aux phosphates/composition chimique , Conformation des protéines , Membrane cellulaire/composition chimique , Membrane cellulaire/immunologie , Caspases initiatrices/composition chimique , Infections bactériennes/immunologie , Bactéries/immunologie
3.
Nature ; 619(7971): 819-827, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-37438530

RÉSUMÉ

Understanding protective immunity to COVID-19 facilitates preparedness for future pandemics and combats new SARS-CoV-2 variants emerging in the human population. Neutralizing antibodies have been widely studied; however, on the basis of large-scale exome sequencing of protected versus severely ill patients with COVID-19, local cell-autonomous defence is also crucial1-4. Here we identify phospholipid scramblase 1 (PLSCR1) as a potent cell-autonomous restriction factor against live SARS-CoV-2 infection in parallel genome-wide CRISPR-Cas9 screens of human lung epithelia and hepatocytes before and after stimulation with interferon-γ (IFNγ). IFNγ-induced PLSCR1 not only restricted SARS-CoV-2 USA-WA1/2020, but was also effective against the Delta B.1.617.2 and Omicron BA.1 lineages. Its robust activity extended to other highly pathogenic coronaviruses, was functionally conserved in bats and mice, and interfered with the uptake of SARS-CoV-2 in both the endocytic and the TMPRSS2-dependent fusion routes. Whole-cell 4Pi single-molecule switching nanoscopy together with bipartite nano-reporter assays found that PLSCR1 directly targeted SARS-CoV-2-containing vesicles to prevent spike-mediated fusion and viral escape. A PLSCR1 C-terminal ß-barrel domain-but not lipid scramblase activity-was essential for this fusogenic blockade. Our mechanistic studies, together with reports that COVID-associated PLSCR1 mutations are found in some susceptible people3,4, identify an anti-coronavirus protein that interferes at a late entry step before viral RNA is released into the host-cell cytosol.


Sujet(s)
COVID-19 , Protéines de transfert des phospholipides , SARS-CoV-2 , Animaux , Humains , Souris , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Chiroptera , COVID-19/immunologie , COVID-19/métabolisme , COVID-19/prévention et contrôle , COVID-19/virologie , , Hépatocytes/immunologie , Hépatocytes/métabolisme , Interféron gamma/immunologie , Poumon/immunologie , Poumon/métabolisme , Fusion membranaire , Protéines de transfert des phospholipides/composition chimique , Protéines de transfert des phospholipides/génétique , Protéines de transfert des phospholipides/immunologie , Protéines de transfert des phospholipides/métabolisme , SARS-CoV-2/classification , SARS-CoV-2/immunologie , SARS-CoV-2/métabolisme , SARS-CoV-2/pathogénicité , Pénétration virale
4.
Sci Signal ; 15(764): eadf0778, 2022 12 13.
Article de Anglais | MEDLINE | ID: mdl-36512642

RÉSUMÉ

Interferons (IFNs) activate cell-autonomous immunity to combat infection and control inflammation. In this issue of Science Signaling, Boccuni et al. reveal how macrophages incorporate stress signals through the p38 MAPK pathway to enhance IFN-induced responses against intracellular pathogens.


Sujet(s)
Interférons , Transduction du signal , p38 Mitogen-Activated Protein Kinases , Macrophages , Logique
5.
Nature ; 607(7918): 339-344, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35768511

RÉSUMÉ

Extreme weather conditions associated with climate change affect many aspects of plant and animal life, including the response to infectious diseases. Production of salicylic acid (SA), a central plant defence hormone1-3, is particularly vulnerable to suppression by short periods of hot weather above the normal plant growth temperature range via an unknown mechanism4-7. Here we show that suppression of SA production in Arabidopsis thaliana at 28 °C is independent of PHYTOCHROME B8,9 (phyB) and EARLY FLOWERING 310 (ELF3), which regulate thermo-responsive plant growth and development. Instead, we found that formation of GUANYLATE BINDING PROTEIN-LIKE 3 (GBPL3) defence-activated biomolecular condensates11 (GDACs) was reduced at the higher growth temperature. The altered GDAC formation in vivo is linked to impaired recruitment of GBPL3 and SA-associated Mediator subunits to the promoters of CBP60g and SARD1, which encode master immune transcription factors. Unlike many other SA signalling components, including the SA receptor and biosynthetic genes, optimized CBP60g expression was sufficient to broadly restore SA production, basal immunity and effector-triggered immunity at the elevated growth temperature without significant growth trade-offs. CBP60g family transcription factors are widely conserved in plants12. These results have implications for safeguarding the plant immune system as well as understanding the concept of the plant-pathogen-environment disease triangle and the emergence of new disease epidemics in a warming climate.


Sujet(s)
Acclimatation , Protéines d'Arabidopsis , Arabidopsis , Environnement , Réchauffement de la planète , Immunité des plantes , Température , Arabidopsis/croissance et développement , Arabidopsis/immunologie , Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Protéines de liaison à la calmoduline/génétique , Régulation de l'expression des gènes végétaux , Réchauffement de la planète/statistiques et données numériques , Interactions hôte-pathogène , Phytochrome B , Maladies des plantes/génétique , Immunité des plantes/génétique , Acide salicylique/métabolisme , Facteurs de transcription
6.
Science ; 373(6552)2021 07 16.
Article de Anglais | MEDLINE | ID: mdl-34437126

RÉSUMÉ

Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.


Sujet(s)
Apolipoprotéines L/métabolisme , Membrane cellulaire/métabolisme , Cytosol/microbiologie , Bactéries à Gram négatif/physiologie , Interféron gamma/immunologie , Apolipoprotéines L/composition chimique , Apolipoprotéines L/génétique , Membrane bactérienne externe/métabolisme , Bactériolyse , Systèmes CRISPR-Cas , Membrane cellulaire/composition chimique , Membrane cellulaire/ultrastructure , Perméabilité des membranes cellulaires , Cellules cultivées , Détergents/métabolisme , Protéines G/métabolisme , Édition de gène , Bactéries à Gram négatif/immunologie , Bactéries à Gram négatif/pathogénicité , Bactéries à Gram négatif/ultrastructure , Humains , Immunité innée , Lipoprotéines/composition chimique , Viabilité microbienne , Antigènes O/métabolisme , Domaines protéiques , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Salmonella typhimurium/physiologie , Salmonella typhimurium/ultrastructure , Solubilité
7.
Nature ; 594(7863): 424-429, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-34040255

RÉSUMÉ

Liquid-liquid phase separation (LLPS) has emerged as a central paradigm for understanding how membraneless organelles compartmentalize diverse cellular activities in eukaryotes1-3. Here we identify a superfamily of plant guanylate-binding protein (GBP)-like GTPases (GBPLs) that assemble LLPS-driven condensates within the nucleus to protect against infection and autoimmunity. In Arabidopsis thaliana, two members of this family-GBPL1 and GBPL3-undergo phase-transition behaviour to control transcriptional responses as part of an allosteric switch that is triggered by exposure to biotic stress. GBPL1, a pseudo-GTPase, sequesters catalytically active GBPL3 under basal conditions but is displaced by GBPL3 LLPS when it enters the nucleus following immune cues to drive the formation of unique membraneless organelles termed GBPL defence-activated condensates (GDACs) that we visualized by in situ cryo-electron tomography. Within these mesoscale GDAC structures, native GBPL3 directly bound defence-gene promoters and recruited specific transcriptional coactivators of the Mediator complex and RNA polymerase II machinery to massively reprogram host gene expression for disease resistance. Together, our study identifies a GBPL circuit that reinforces the biological importance of phase-separated condensates, in this case, as indispensable players in plant defence.


Sujet(s)
Arabidopsis/immunologie , Noyau de la cellule/composition chimique , Noyau de la cellule/métabolisme , Protéines G/métabolisme , Protéines intrinsèquement désordonnées/métabolisme , Transition de phase , Immunité des plantes , Arabidopsis/génétique , Arabidopsis/métabolisme , Arabidopsis/ultrastructure , Noyau de la cellule/génétique , Noyau de la cellule/ultrastructure , Chromatine/génétique , Cryomicroscopie électronique , Protéines G/composition chimique , Protéines G/ultrastructure , Régulation de l'expression des gènes végétaux/génétique , Protéines intrinsèquement désordonnées/composition chimique , Protéines intrinsèquement désordonnées/ultrastructure , Complexe médiateur , Famille multigénique/génétique , Organites/composition chimique , Organites/immunologie , Organites/métabolisme , Organites/ultrastructure , Cellules végétales/composition chimique , Cellules végétales/immunologie , Cellules végétales/métabolisme , Cellules végétales/ultrastructure , Maladies des plantes/immunologie , Immunité des plantes/génétique , Régions promotrices (génétique)/génétique , RNA polymerase II/métabolisme , Transcription génétique
8.
Nat Immunol ; 21(8): 880-891, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32541830

RÉSUMÉ

Bacterial lipopolysaccharide triggers human caspase-4 (murine caspase-11) to cleave gasdermin-D and induce pyroptotic cell death. How lipopolysaccharide sequestered in the membranes of cytosol-invading bacteria activates caspases remains unknown. Here we show that in interferon-γ-stimulated cells guanylate-binding proteins (GBPs) assemble on the surface of Gram-negative bacteria into polyvalent signaling platforms required for activation of caspase-4. Caspase-4 activation is hierarchically controlled by GBPs; GBP1 initiates platform assembly, GBP2 and GBP4 control caspase-4 recruitment, and GBP3 governs caspase-4 activation. In response to cytosol-invading bacteria, activation of caspase-4 through the GBP platform is essential to induce gasdermin-D-dependent pyroptosis and processing of interleukin-18, thereby destroying the replicative niche for intracellular bacteria and alerting neighboring cells, respectively. Caspase-11 and GBPs epistatically protect mice against lethal bacterial challenge. Multiple antagonists of the pathway encoded by Shigella flexneri, a cytosol-adapted bacterium, provide compelling evolutionary evidence for the importance of the GBP-caspase-4 pathway in antibacterial defense.


Sujet(s)
Caspases initiatrices/immunologie , Protéines G/immunologie , Infections bactériennes à Gram négatif/immunologie , Inflammasomes/immunologie , Transduction du signal/immunologie , Animaux , Bactéries à Gram négatif/immunologie , Cellules HeLa , Humains , Lipopolysaccharides/immunologie , Souris , Pyroptose/immunologie
9.
Curr Opin Immunol ; 60: 71-80, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31176142

RÉSUMÉ

Inside host cells, guanylate binding proteins (GBPs) rapidly assemble into large antimicrobial defense complexes that combat a wide variety of bacterial pathogens. These massive nanomachines often completely coat targeted microbes where they act as recruitment platforms for downstream effectors capable of direct bactericidal activity. GBP-containing platforms also serve as sensory hubs to activate inflammasome-driven responses in the mammalian cytosol while in plants like Arabidopsis, GBP orthologues may facilitate intranuclear signaling for immunity against invasive phytopathogens. Together, this group of immune GTPases serve as a major defensive repertoire to protect the host cell interior from bacterial colonization across plant and animal kingdoms.


Sujet(s)
Cellules eucaryotes/immunologie , Cellules eucaryotes/métabolisme , Protéines G/métabolisme , Interactions hôte-pathogène/immunologie , Immunité , Interférons/métabolisme , Animaux , Infections bactériennes/génétique , Infections bactériennes/immunologie , Infections bactériennes/métabolisme , Infections bactériennes/microbiologie , Évolution biologique , Cellules eucaryotes/microbiologie , Protéines G/composition chimique , Protéines G/génétique , Régulation de l'expression des gènes , Régulation de l'expression des gènes codant pour des enzymes , Humains , Maladies des plantes , Phénomènes physiologiques des plantes , Plantes/génétique , Plantes/immunologie , Plantes/métabolisme
10.
J Exp Med ; 216(3): 482-500, 2019 03 04.
Article de Anglais | MEDLINE | ID: mdl-30755454

RÉSUMÉ

Guanylate-binding proteins (GBPs) have recently emerged as central orchestrators of immunity to infection, inflammation, and neoplastic diseases. Within numerous host cell types, these IFN-induced GTPases assemble into large nanomachines that execute distinct host defense activities against a wide variety of microbial pathogens. In addition, GBPs customize inflammasome responses to bacterial infection and sepsis, where they act as critical rheostats to amplify innate immunity and regulate tissue damage. Similar functions are becoming evident for metabolic inflammatory syndromes and cancer, further underscoring the importance of GBPs within infectious as well as altered homeostatic settings. A better understanding of the basic biology of these IFN-induced GTPases could thus benefit clinical approaches to a wide spectrum of important human diseases.


Sujet(s)
Protéines G/composition chimique , Protéines G/métabolisme , Interactions hôte-parasite/immunologie , Interactions hôte-pathogène/immunologie , Interférons/métabolisme , Animaux , Colite/immunologie , Colite/métabolisme , Protéines G/immunologie , Humains , Inflammasomes/physiologie , Inflammation/immunologie , Inflammation/métabolisme , Vertébrés
11.
mBio ; 9(4)2018 08 28.
Article de Anglais | MEDLINE | ID: mdl-30154263

RÉSUMÉ

Phagocytic cells are the first line of innate defense against intracellular pathogens, and yet Toxoplasma gondii is renowned for its ability to survive in macrophages, although this paradigm is based on virulent type I parasites. Surprisingly, we find that avirulent type III parasites are preferentially cleared in naive macrophages, independent of gamma interferon (IFN-γ) activation. The ability of naive macrophages to clear type III parasites was dependent on enhanced activity of NADPH oxidase (Nox)-generated reactive oxygen species (ROS) and induction of guanylate binding protein 5 (Gbp5). Macrophages infected with type III parasites (CTG strain) showed a time-dependent increase in intracellular ROS generation that was higher than that induced by type I parasites (GT1 strain). The absence of Nox1 or Nox2, gp91 subunit isoforms of the Nox complex, reversed ROS-mediated clearance of CTG parasites. Consistent with this finding, both Nox1-/- and Nox2-/- mice showed higher susceptibility to CTG infection than wild-type mice. Additionally, Gbp5 expression was induced upon infection and the enhanced clearance of CTG strain parasites was reversed in Gbp5-/- macrophages. Expression of a type I ROP18 allele in CTG prevented clearance in naive macrophages, suggesting that it plays a role counteracting Gbp5. Although ROS and Gbp5 have been linked to activation of the NLRP3 inflammasome, clearance of CTG parasites did not rely on induction of pyroptosis. Collectively, these findings reveal that not all strains of T. gondii are adept at avoiding clearance in macrophages and define new roles for ROS and Gbps in controlling this important intracellular pathogen.IMPORTANCEToxoplasma infections in humans and other mammals are largely controlled by IFN-γ produced by the activated adaptive immune system. However, we still do not completely understand the role of cell-intrinsic functions in controlling Toxoplasma or other apicomplexan infections. The present work identifies intrinsic activities in naive macrophages in counteracting T. gondii infection. Using an avirulent strain of T. gondii, we highlight the importance of Nox complexes in conferring protection against parasite infection both in vitro and in vivo We also identify Gbp5 as a novel macrophage factor involved in limiting intracellular infection by avirulent strains of T. gondii The rarity of human infections caused by type III strains suggests that these mechanisms may also be important in controlling human toxoplasmosis. These findings further extend our understanding of host responses and defense mechanisms that act to control parasitic infections at the cellular level.


Sujet(s)
Protéines G/métabolisme , Macrophages/parasitologie , NADPH Oxidase 1/métabolisme , NADPH Oxidase 2/métabolisme , Toxoplasmose/immunologie , Animaux , Cellules cultivées , Protéines G/génétique , Immunité innée , Interférons/immunologie , Macrophages/immunologie , Souris , Souris de lignée C57BL , Souris knockout , NADPH Oxidase 1/génétique , NADPH Oxidase 2/génétique , Cellules RAW 264.7 , Espèces réactives de l'oxygène/métabolisme , Toxoplasma , Virulence
12.
Nature ; 551(7680): 303-305, 2017 11 16.
Article de Anglais | MEDLINE | ID: mdl-29072295
14.
Nat Commun ; 8: 15865, 2017 06 28.
Article de Anglais | MEDLINE | ID: mdl-28656966

RÉSUMÉ

Optimal regulation of the innate immune receptor nucleotide-binding oligomerization domain-containing protein 2 (NOD2) is essential for controlling bacterial infections and inflammatory disorders. Chronic NOD2 stimulation induces non-responsiveness to restimulation, termed NOD2-induced tolerance. Although the levels of the NOD2 adaptor, RIP2, are reported to regulate both acute and chronic NOD2 signalling, how RIP2 levels are modulated is unclear. Here we show that ZNRF4 induces K48-linked ubiquitination of RIP2 and promotes RIP2 degradation. A fraction of RIP2 localizes to the endoplasmic reticulum (ER), where it interacts with ZNRF4 under either 55 unstimulated and muramyl dipeptide-stimulated conditions. Znrf4 knockdown monocytes have sustained nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation, and Znrf4 knockdown mice have reduced NOD2-induced tolerance and more effective control of Listeria monocytogenes infection. Our results thus demonstrate E3-ubiquitin ligase ZNRF4-mediated RIP2 degradation as a negative regulatory mechanism of NOD2-induced NF-κB, cytokine and anti-bacterial responses in vitro and in vivo, and identify a ZNRF4-RIP2 axis of fine-tuning NOD2 signalling to promote protective host immunity.


Sujet(s)
Acétylmuramyl alanyl isoglutamine/pharmacologie , Protéines de liaison à l'ADN/métabolisme , Tolérance immunitaire , Protéine adaptatrice de signalisation NOD2/métabolisme , Acétylmuramyl alanyl isoglutamine/immunologie , Animaux , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Cellules HEK293 , Humains , Tolérance immunitaire/effets des médicaments et des substances chimiques , Listeria monocytogenes/pathogénicité , Infections à Listeria/immunologie , Infections à Listeria/métabolisme , Souris de lignée C57BL , Souches mutantes de souris , Monocytes/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Receptor-Interacting Protein Serine-Threonine Kinase 2/métabolisme , Transduction du signal/physiologie , Ubiquitination/effets des médicaments et des substances chimiques
15.
Nat Immunol ; 17(5): 481-9, 2016 May.
Article de Anglais | MEDLINE | ID: mdl-27092805

RÉSUMÉ

Traditional views of the inflammasome highlight the assembly of pre-existing core components shortly after infection or tissue damage. Emerging work, however, suggests that the inflammasome machinery is also subject to 'tunable' or inducible signals that might accelerate its autocatalytic properties and dictate where inflammasome assembly takes place in the cell. Many of these signals operate downstream of interferon receptors to elicit inflammasome regulators, including a new family of interferon-induced GTPases called 'guanylate-binding proteins' (GBPs). Here we investigate the critical roles of interferon-induced GBPs in directing inflammasome subtype-specific responses and their consequences for cell-autonomous immunity to a wide variety of microbial pathogens. We discuss emerging mechanisms of action and the potential effect of these GBPs on predisposition to sepsis and other infectious or inflammatory diseases.


Sujet(s)
Protéines G/immunologie , Inflammasomes/immunologie , Interférons/immunologie , Transduction du signal/immunologie , Animaux , Résistance à la maladie/génétique , Résistance à la maladie/immunologie , Protéines G/classification , Protéines G/génétique , Interactions hôte-pathogène/immunologie , Humains , Infections/immunologie , Infections/microbiologie , Infections/parasitologie , Inflammasomes/génétique , Inflammasomes/métabolisme , Interférons/métabolisme , Listeria monocytogenes/immunologie , Listeria monocytogenes/physiologie , Souris , Modèles immunologiques , Phylogenèse , Transduction du signal/génétique , Toxoplasma/immunologie , Toxoplasma/physiologie
16.
Microbiol Spectr ; 4(6)2016 12.
Article de Anglais | MEDLINE | ID: mdl-28087931

RÉSUMÉ

Specialized adaptations for killing microbes are synonymous with phagocytic cells including macrophages, monocytes, inflammatory neutrophils, and eosinophils. Recent genome sequencing of extant species, however, reveals that analogous antimicrobial machineries exist in certain non-immune cells and also within species that ostensibly lack a well-defined immune system. Here we probe the evolutionary record for clues about the ancient and diverse phylogenetic origins of macrophage killing mechanisms and how some of their properties are shared with cells outside the traditional bounds of immunity in higher vertebrates such as mammals.


Sujet(s)
Évolution biologique , Macrophages/immunologie , Viabilité microbienne , Phagocytose , Animaux , Humains
17.
J Biol Chem ; 291(3): 1123-36, 2016 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-26555265

RÉSUMÉ

Many immunostimulants act as vaccine adjuvants via activation of the innate immune system, although in many cases it is unclear which specific molecules contribute to the stimulatory activity. QS-21 is a defined, highly purified, and soluble saponin adjuvant currently used in licensed and exploratory vaccines, including vaccines against malaria, cancer, and HIV-1. However, little is known about the mechanisms of cellular activation induced by QS-21. We observed QS-21 to elicit caspase-1-dependent IL-1ß and IL-18 release in antigen-presenting cells such as macrophages and dendritic cells when co-stimulated with the TLR4-agonist adjuvant monophosphoryl lipid A. Furthermore, our data suggest that the ASC-NLRP3 inflammasome is responsible for QS-21-induced IL-1ß/IL-18 release. At higher concentrations, QS-21 induced macrophage and dendritic cell death in a caspase-1-, ASC-, and NLRP3-independent manner, whereas the presence of cholesterol rescued cell viability. A nanoparticulate adjuvant that contains QS-21 as part of a heterogeneous mixture of saponins also induced IL-1ß in an NLRP3-dependent manner. Interestingly, despite the role NLRP3 plays for cellular activation in vitro, NLRP3-deficient mice immunized with HIV-1 gp120 and QS-21 showed significantly higher levels of Th1 and Th2 antigen-specific T cell responses and increased IgG1 and IgG2c compared with wild type controls. Thus, we have identified QS-21 as a nonparticulate single molecular saponin that activates the NLRP3 inflammasome, but this signaling pathway may contribute to decreased antigen-specific responses in vivo.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Protéines de transport/métabolisme , Cellules dendritiques/effets des médicaments et des substances chimiques , Immunité innée/effets des médicaments et des substances chimiques , Inflammasomes/effets des médicaments et des substances chimiques , Macrophages/effets des médicaments et des substances chimiques , Saponines/pharmacologie , Vaccins contre le SIDA/agonistes , Vaccins contre le SIDA/immunologie , Adjuvants immunologiques/analyse , Adjuvants immunologiques/composition chimique , Animaux , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/immunologie , Protéines de transport/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Protéines G/génétique , Protéines G/métabolisme , Protéine d'enveloppe gp120 du VIH/agonistes , Protéine d'enveloppe gp120 du VIH/immunologie , Immunoglobuline G/analyse , Immunoglobuline G/biosynthèse , Inflammasomes/immunologie , Inflammasomes/métabolisme , Lipide A/agonistes , Lipide A/analogues et dérivés , Lipide A/pharmacologie , Macrophages/cytologie , Macrophages/immunologie , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine , Saponines/analyse , Saponines/composition chimique , Solubilité , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/métabolisme
18.
Article de Anglais | MEDLINE | ID: mdl-25081628

RÉSUMÉ

Few pathogens run the gauntlet of sterilizing immunity like Mycobacterium tuberculosis (Mtb). This organism infects mononuclear phagocytes and is also ingested by neutrophils, both of which possess an arsenal of cell-intrinsic effector mechanisms capable of eliminating it. Here Mtb encounters acid, oxidants, nitrosylating agents, and redox congeners, often exuberantly delivered under low oxygen tension. Further pressure is applied by withholding divalent Fe²âº, Mn²âº, Cu²âº, and Zn²âº, as well as by metabolic privation in the form of carbon needed for anaplerosis and aromatic amino acids for growth. Finally, host E3 ligases ubiquinate, cationic peptides disrupt, and lysosomal enzymes digest Mtb as part of the autophagic response to this particular pathogen. It is a testament to the evolutionary fitness of Mtb that sterilization is rarely complete, although sufficient to ensure most people infected with this airborne bacterium remain disease-free.


Sujet(s)
Immunité innée , Macrophages/immunologie , Mycobacterium tuberculosis/immunologie , Phagocytes/immunologie , Acides aminés/métabolisme , Peptides antimicrobiens cationiques , Carbone/métabolisme , Cations/métabolisme , Humains , Concentration en ions d'hydrogène , Fer/métabolisme , Lysosomes , Stress oxydatif/immunologie , Espèces réactives de l'oxygène , Récepteur calcitriol/immunologie , Récepteur interféron/immunologie , Récepteurs à l'interleukine-1/immunologie , Récepteurs aux facteurs de nécrose tumorale/immunologie , Récepteurs de type Toll/immunologie
19.
PLoS Pathog ; 9(7): e1003491, 2013.
Article de Anglais | MEDLINE | ID: mdl-23853600

RÉSUMÉ

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Sujet(s)
Encéphale/immunologie , Régulation de l'expression des gènes , Immunité innée , Facteurs de régulation d'interféron/métabolisme , Paludisme cérébral/immunologie , Protéines de tissu nerveux/métabolisme , Plasmodium berghei/immunologie , Substitution d'acide aminé , Animaux , Sites de fixation , Encéphale/métabolisme , Encéphale/parasitologie , Cellules cultivées , Cytokines/biosynthèse , Cytokines/sang , Analyse de profil d'expression de gènes , Facteur-1 de régulation d'interféron/génétique , Facteur-1 de régulation d'interféron/métabolisme , Facteurs de régulation d'interféron/composition chimique , Facteurs de régulation d'interféron/génétique , Paludisme cérébral/sang , Paludisme cérébral/métabolisme , Paludisme cérébral/parasitologie , Souris , Souris knockout , Souches mutantes de souris , Protéines mutantes/composition chimique , Protéines mutantes/métabolisme , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/génétique , Neurones/immunologie , Neurones/métabolisme , Neurones/parasitologie , Organismes exempts d'organismes pathogènes spécifiques , Rate/immunologie , Rate/métabolisme , Rate/anatomopathologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Lymphocytes auxiliaires Th1/parasitologie
20.
Proc Natl Acad Sci U S A ; 110(32): E2997-3006, 2013 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-23882081

RÉSUMÉ

Macrophage migration inhibitory factor (MIF), an innate cytokine encoded in a functionally polymorphic genetic locus, contributes to detrimental inflammation but may be crucial for controlling infection. We explored the role of variant MIF alleles in tuberculosis. In a Ugandan cohort, genetic low expressers of MIF were 2.4-times more frequently identified among patients with Mycobacterium tuberculosis (TB) bacteremia than those without. We also found mycobacteria-stimulated transcription of MIF and serum MIF levels to be correlated with MIF genotype in human macrophages and in a separate cohort of US TB patients, respectively. To determine mechanisms for MIF's protective role, we studied both aerosolized and i.v. models of mycobacterial infection and observed MIF-deficient mice to succumb more quickly with higher organism burden, increased lung pathology, and decreased innate cytokine production (TNF-α, IL-12, IL-10). MIF-deficient animals showed increased pulmonary neutrophil accumulation but preserved adaptive immune response. MIF-deficient macrophages demonstrated decreased cytokine and reactive oxygen production and impaired mycobacterial killing. Transcriptional investigation of MIF-deficient macrophages revealed reduced expression of the pattern recognition receptor dectin-1; restoration of dectin-1 expression recovered innate cytokine production and mycobacterial killing. Our data place MIF in a crucial upstream position in the innate immune response to mycobacteria and suggest that commonly occurring low expression MIF alleles confer an increased risk of TB disease in some populations.


Sujet(s)
Immunité innée/immunologie , Facteurs inhibiteurs de la migration des macrophages/immunologie , Mycobacterium tuberculosis/immunologie , Tuberculose/immunologie , Adulte , Animaux , Lignée cellulaire , Cytokines/immunologie , Cytokines/métabolisme , Femelle , Expression des gènes/immunologie , Génotype , Humains , Immunité innée/génétique , Lectines de type C/génétique , Lectines de type C/immunologie , Lectines de type C/métabolisme , Poumon/immunologie , Poumon/métabolisme , Poumon/microbiologie , Facteurs inhibiteurs de la migration des macrophages/sang , Facteurs inhibiteurs de la migration des macrophages/génétique , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/microbiologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Polymorphisme génétique , RT-PCR , Taux de survie , Tuberculose/génétique , Tuberculose/mortalité , Ouganda , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...