Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres











Base de données
Gamme d'année
2.
Immunity ; 56(8): 1701-1703, 2023 08 08.
Article de Anglais | MEDLINE | ID: mdl-37557076

RÉSUMÉ

Whether soluble TREM2 has a functional role in the central nervous system has been unclear. In this issue of Immunity, Zhong et al. show that soluble TREM2 inhibits aberrant synaptic pruning by sopping up the complement factor C1q to protect neurons and mitigate neurodegeneration.


Sujet(s)
Microglie , Synapses , Protéines du système du complément , Neurones , Complément C1q
4.
EMBO J ; 41(4): e109108, 2022 02 15.
Article de Anglais | MEDLINE | ID: mdl-35019161

RÉSUMÉ

Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.


Sujet(s)
Dégénérescence lobaire frontotemporale/anatomopathologie , Glycoprotéines membranaires/métabolisme , Microglie/physiologie , Monocytes/métabolisme , Progranulines/déficit , Récepteurs immunologiques/métabolisme , Animaux , Anticorps/immunologie , Anticorps/pharmacologie , Encéphale/imagerie diagnostique , Encéphale/physiopathologie , Modèles animaux de maladie humaine , Femelle , Dégénérescence lobaire frontotemporale/métabolisme , Humains , Lysosomes/métabolisme , Lysosomes/anatomopathologie , Mâle , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/immunologie , Souris de lignée C57BL , Souris knockout , Microglie/effets des médicaments et des substances chimiques , Monocytes/effets des médicaments et des substances chimiques , Récepteurs immunologiques/génétique , Récepteurs immunologiques/immunologie , Syk kinase/métabolisme
5.
Cell ; 184(18): 4651-4668.e25, 2021 09 02.
Article de Anglais | MEDLINE | ID: mdl-34450028

RÉSUMÉ

GRN mutations cause frontotemporal dementia (GRN-FTD) due to deficiency in progranulin (PGRN), a lysosomal and secreted protein with unclear function. Here, we found that Grn-/- mice exhibit a global deficiency in bis(monoacylglycero)phosphate (BMP), an endolysosomal phospholipid we identified as a pH-dependent PGRN interactor as well as a redox-sensitive enhancer of lysosomal proteolysis and lipolysis. Grn-/- brains also showed an age-dependent, secondary storage of glucocerebrosidase substrate glucosylsphingosine. We investigated a protein replacement strategy by engineering protein transport vehicle (PTV):PGRN-a recombinant protein linking PGRN to a modified Fc domain that binds human transferrin receptor for enhanced CNS biodistribution. PTV:PGRN rescued various Grn-/- phenotypes in primary murine macrophages and human iPSC-derived microglia, including oxidative stress, lysosomal dysfunction, and endomembrane damage. Peripherally delivered PTV:PGRN corrected levels of BMP, glucosylsphingosine, and disease pathology in Grn-/- CNS, including microgliosis, lipofuscinosis, and neuronal damage. PTV:PGRN thus represents a potential biotherapeutic for GRN-FTD.


Sujet(s)
Produits biologiques/usage thérapeutique , Encéphale/métabolisme , Maladies lysosomiales/thérapie , Progranulines/usage thérapeutique , Animaux , Protéines morphogénétiques osseuses/métabolisme , Endosomes/métabolisme , Femelle , Démence frontotemporale/sang , Démence frontotemporale/liquide cérébrospinal , Gliose/complications , Gliose/anatomopathologie , Humains , Cellules souches pluripotentes induites/métabolisme , Inflammation/anatomopathologie , Métabolisme lipidique , Lipofuscine/métabolisme , Lysosomes/métabolisme , Macrophages/métabolisme , Mâle , Glycoprotéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , Microglie/métabolisme , Dégénérescence nerveuse/anatomopathologie , Phénotype , Progranulines/déficit , Progranulines/métabolisme , Récepteurs immunologiques/métabolisme , Récepteurs à la transferrine/métabolisme , Distribution tissulaire
7.
Nat Metab ; 3(2): 211-227, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33619376

RÉSUMÉ

Proregenerative responses are required for the restoration of nervous-system functionality in demyelinating diseases such as multiple sclerosis (MS). Yet, the limiting factors responsible for poor CNS repair are only partially understood. Here, we test the impact of a Western diet (WD) on phagocyte function in a mouse model of demyelinating injury that requires microglial innate immune function for a regenerative response to occur. We find that WD feeding triggers an ageing-related, dysfunctional metabolic response that is associated with impaired myelin-debris clearance in microglia, thereby impairing lesion recovery after demyelination. Mechanistically, we detect enhanced transforming growth factor beta (TGFß) signalling, which suppresses the activation of the liver X receptor (LXR)-regulated genes involved in cholesterol efflux, thereby inhibiting phagocytic clearance of myelin and cholesterol. Blocking TGFß or promoting triggering receptor expressed on myeloid cells 2 (TREM2) activity restores microglia responsiveness and myelin-debris clearance after demyelinating injury. Thus, we have identified a druggable microglial immune checkpoint mechanism regulating the microglial response to injury that promotes remyelination.


Sujet(s)
Maladies démyélinisantes/immunologie , Maladies démyélinisantes/métabolisme , Régime alimentaire , Immunité innée/immunologie , Facteur de croissance transformant bêta/métabolisme , Vieillissement/métabolisme , Animaux , Cholestérol/métabolisme , Régime occidental , Récepteurs hépatiques X , Lysolécithine/pharmacologie , Glycoprotéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Microglie/métabolisme , Gaine de myéline/métabolisme , Phagocytes/métabolisme , Récepteurs immunologiques/métabolisme
8.
Neuron ; 108(5): 801-821, 2020 12 09.
Article de Anglais | MEDLINE | ID: mdl-33096024

RÉSUMÉ

Alzheimer's disease (AD) is currently untreatable, and therapeutic strategies aimed to slow cognitive decline have not yet been successful. Many of these approaches have targeted the amyloid cascade, indicating that novel treatment strategies are required. Recent genome-wide association studies (GWASs) have identified a number of risk factors in genes expressed in microglia, underscoring their therapeutic potential in neurodegeneration. In this review, we discuss how the recently defined functions of these AD risk genes can be targeted therapeutically to modulate microglial cell state and slow the progression of AD. Antibody-mediated stimulation of the triggering receptor of myeloid cells 2 (TREM2) is on the forefront of these candidate therapeutic approaches based on a combination of compelling human genetics and emerging preclinical data. This and other approaches to modify microglial function are a topic of intensive study and provide an opportunity for innovative AD treatments, which may be applied alone or potentially in combination with classical anti-amyloid therapies.


Sujet(s)
Maladie d'Alzheimer/immunologie , Maladie d'Alzheimer/thérapie , Encéphale/immunologie , Thérapie génétique/tendances , Immunothérapie/tendances , Microglie/immunologie , Maladie d'Alzheimer/génétique , Animaux , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Encéphale/effets des médicaments et des substances chimiques , Étude d'association pangénomique/tendances , Humains , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/immunologie , Glycoprotéines membranaires/usage thérapeutique , Microglie/effets des médicaments et des substances chimiques , Récepteurs immunologiques/génétique , Récepteurs immunologiques/immunologie , Récepteurs immunologiques/usage thérapeutique
9.
Nat Neurosci ; 23(8): 927-938, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32514138

RÉSUMÉ

Human genetic data indicate that microglial dysfunction contributes to the pathology of Alzheimer's disease (AD), exemplified by the identification of coding variants in triggering receptor expressed on myeloid cells 2 (TREM2) and, more recently, in PLCG2, a phospholipase-encoding gene expressed in microglia. Although studies in mouse models have implicated specific Trem2-dependent microglial functions in AD, the underlying molecular mechanisms and translatability to human disease remain poorly defined. In this study, we used genetically engineered human induced pluripotent stem cell-derived microglia-like cells to show that TREM2 signals through PLCγ2 to mediate cell survival, phagocytosis, processing of neuronal debris, and lipid metabolism. Loss of TREM2 or PLCγ2 signaling leads to a shared signature of transcriptional dysregulation that underlies these phenotypes. Independent of TREM2, PLCγ2 also signals downstream of Toll-like receptors to mediate inflammatory responses. Therefore, PLCγ2 activity regulates divergent microglial functions via distinct TREM2-dependent and -independent signaling and might be involved in the transition to a microglial state associated with neurodegenerative disease.


Sujet(s)
Inflammation/métabolisme , Glycoprotéines membranaires/métabolisme , Microglie/métabolisme , Phospholipase C gamma/métabolisme , Récepteurs immunologiques/métabolisme , Transduction du signal/physiologie , Animaux , Survie cellulaire/physiologie , Humains , Cellules souches pluripotentes induites/métabolisme , Glycoprotéines membranaires/génétique , Souris , Souris knockout , Neurones/métabolisme , Phagocytose/physiologie , Phospholipase C gamma/génétique , Récepteurs immunologiques/génétique
10.
EMBO Mol Med ; 12(4): e11227, 2020 04 07.
Article de Anglais | MEDLINE | ID: mdl-32154671

RÉSUMÉ

Triggering receptor expressed on myeloid cells 2 (TREM2) is essential for the transition of homeostatic microglia to a disease-associated microglial state. To enhance TREM2 activity, we sought to selectively increase the full-length protein on the cell surface via reducing its proteolytic shedding by A Disintegrin And Metalloproteinase (i.e., α-secretase) 10/17. We screened a panel of monoclonal antibodies against TREM2, with the aim to selectively compete for α-secretase-mediated shedding. Monoclonal antibody 4D9, which has a stalk region epitope close to the cleavage site, demonstrated dual mechanisms of action by stabilizing TREM2 on the cell surface and reducing its shedding, and concomitantly activating phospho-SYK signaling. 4D9 stimulated survival of macrophages and increased microglial uptake of myelin debris and amyloid ß-peptide in vitro. In vivo target engagement was demonstrated in cerebrospinal fluid, where nearly all soluble TREM2 was 4D9-bound. Moreover, in a mouse model for Alzheimer's disease-related pathology, 4D9 reduced amyloidogenesis, enhanced microglial TREM2 expression, and reduced a homeostatic marker, suggesting a protective function by driving microglia toward a disease-associated state.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Glycoprotéines membranaires/immunologie , Microglie , Myélome multiple , Récepteurs immunologiques/immunologie , Peptides bêta-amyloïdes , Animaux , Lignée cellulaire tumorale , Femelle , Macrophages , Souris , Microglie/anatomopathologie , Rats , Rat Wistar
11.
Neuron ; 105(5): 837-854.e9, 2020 03 04.
Article de Anglais | MEDLINE | ID: mdl-31902528

RÉSUMÉ

Loss-of-function (LOF) variants of TREM2, an immune receptor expressed in microglia, increase Alzheimer's disease risk. TREM2 senses lipids and mediates myelin phagocytosis, but its role in microglial lipid metabolism is unknown. Combining chronic demyelination paradigms and cell sorting with RNA sequencing and lipidomics, we find that wild-type microglia acquire a disease-associated transcriptional state, while TREM2-deficient microglia remain largely homeostatic, leading to neuronal damage. TREM2-deficient microglia phagocytose myelin debris but fail to clear myelin cholesterol, resulting in cholesteryl ester (CE) accumulation. CE increase is also observed in APOE-deficient glial cells, reflecting impaired brain cholesterol transport. This finding replicates in myelin-treated TREM2-deficient murine macrophages and human iPSC-derived microglia, where it is rescued by an ACAT1 inhibitor and LXR agonist. Our studies identify TREM2 as a key transcriptional regulator of cholesterol transport and metabolism under conditions of chronic myelin phagocytic activity, as TREM2 LOF causes pathogenic lipid accumulation in microglia.


Sujet(s)
Encéphale/métabolisme , Cholestérol/métabolisme , Macrophages/métabolisme , Glycoprotéines membranaires/génétique , Microglie/métabolisme , Gaine de myéline/métabolisme , Phagocytose/génétique , Récepteurs immunologiques/génétique , Acetyl-coA C-acetyltransferase/antagonistes et inhibiteurs , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Animaux , Cholestérol ester/métabolisme , Modèles animaux de maladie humaine , Cytométrie en flux , Humains , Cellules souches pluripotentes induites , Métabolisme lipidique/génétique , Lipidomique , Récepteurs hépatiques X/agonistes , Souris , Souris knockout , Souris invalidées pour les gènes ApoE , RNA-Seq
13.
Cell Rep ; 12(10): 1555-1563, 2015 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-26321639

RÉSUMÉ

The progressive depletion of CD4 T cells underlies clinical progression to AIDS in untreated HIV-infected subjects. Most dying CD4 T cells correspond to resting nonpermissive cells residing in lymphoid tissues. Death is due to an innate immune response against the incomplete cytosolic viral DNA intermediates accumulating in these cells. The viral DNA is detected by the IFI16 sensor, leading to inflammasome assembly, caspase-1 activation, and the induction of pyroptosis, a highly inflammatory form of programmed cell death. We now show that cell-to-cell transmission of HIV is obligatorily required for activation of this death pathway. Cell-free HIV-1 virions, even when added in large quantities, fail to activate pyroptosis. These findings underscore the infected CD4 T cells as the major killing units promoting progression to AIDS and highlight a previously unappreciated role for the virological synapse in HIV pathogenesis.


Sujet(s)
Lymphocytes T CD4+/physiologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Apoptose , Lymphocytes T CD4+/virologie , Caspase-1/métabolisme , Molécules d'adhérence cellulaire/physiologie , Cellules cultivées , Techniques de coculture , Infections à VIH/transmission , Humains , Tissu lymphoïde/anatomopathologie
14.
Science ; 343(6169): 428-32, 2014 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-24356113

RÉSUMÉ

The progressive depletion of quiescent "bystander" CD4 T cells, which are nonpermissive to HIV infection, is a principal driver of the acquired immunodeficiency syndrome (AIDS). These cells undergo abortive infection characterized by the cytosolic accumulation of incomplete HIV reverse transcripts. These viral DNAs are sensed by an unidentified host sensor that triggers an innate immune response, leading to caspase-1 activation and pyroptosis. Using unbiased proteomic and targeted biochemical approaches, as well as two independent methods of lentiviral short hairpin RNA-mediated gene knockdown in primary CD4 T cells, we identify interferon-γ-inducible protein 16 (IFI16) as a host DNA sensor required for CD4 T cell death due to abortive HIV infection. These findings provide insights into a key host pathway that plays a central role in CD4 T cell depletion during disease progression to AIDS.


Sujet(s)
Syndrome d'immunodéficience acquise/immunologie , Apoptose/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/virologie , ADN viral/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Protéines nucléaires/immunologie , Phosphoprotéines/immunologie , Cellules cultivées , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Techniques de knock-down de gènes , Cellules HEK293 , Humains , Immunité innée , Déplétion lymphocytaire , Protéines nucléaires/génétique , Tonsille palatine/immunologie , Phosphoprotéines/génétique , Petit ARN interférent/génétique , Rate/immunologie
15.
Nature ; 505(7484): 509-14, 2014 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-24356306

RÉSUMÉ

The pathway causing CD4 T-cell death in HIV-infected hosts remains poorly understood although apoptosis has been proposed as a key mechanism. We now show that caspase-3-mediated apoptosis accounts for the death of only a small fraction of CD4 T cells corresponding to those that are both activated and productively infected. The remaining over 95% of quiescent lymphoid CD4 T cells die by caspase-1-mediated pyroptosis triggered by abortive viral infection. Pyroptosis corresponds to an intensely inflammatory form of programmed cell death in which cytoplasmic contents and pro-inflammatory cytokines, including IL-1ß, are released. This death pathway thus links the two signature events in HIV infection-CD4 T-cell depletion and chronic inflammation-and creates a pathogenic vicious cycle in which dying CD4 T cells release inflammatory signals that attract more cells to die. This cycle can be broken by caspase 1 inhibitors shown to be safe in humans, raising the possibility of a new class of 'anti-AIDS' therapeutics targeting the host rather than the virus.


Sujet(s)
Lymphocytes T CD4+/anatomopathologie , Caspase-1/métabolisme , Infections à VIH/immunologie , Infections à VIH/anatomopathologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/pathogénicité , Administration par voie orale , Adulte , Agents antiVIH/pharmacologie , Lymphocytes T CD4+/cytologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/métabolisme , Caspase-3/métabolisme , Inhibiteurs des caspases/administration et posologie , Inhibiteurs des caspases/pharmacologie , Mort cellulaire/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Infections à VIH/enzymologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , Humains , Techniques in vitro , Inflammasomes/immunologie , Inflammasomes/métabolisme , Inflammation/complications , Inflammation/immunologie , Inflammation/anatomopathologie , Inflammation/virologie , Interleukine-1 bêta/biosynthèse , Interleukine-1 bêta/métabolisme , Noeuds lymphatiques/enzymologie , Mâle , Tonsille palatine/effets des médicaments et des substances chimiques , Tonsille palatine/virologie , Précurseurs de protéines/biosynthèse , Rate/effets des médicaments et des substances chimiques , Rate/virologie , Réplication virale
16.
Dev Biol ; 381(2): 434-45, 2013 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-23796903

RÉSUMÉ

The evolutionarily conserved JNK/AP-1 (Jun N-terminal kinase/activator protein 1) and BMP (Bone Morphogenetic Protein) signaling cascades are deployed hierarchically to regulate dorsal closure in the fruit fly Drosophila melanogaster. In this developmental context, the JNK/AP-1 signaling cascade transcriptionally activates BMP signaling in leading edge epidermal cells. Here we show that the mummy (mmy) gene product, which is required for dorsal closure, functions as a BMP signaling antagonist. Genetic and biochemical tests of Mmy's role as a BMP-antagonist indicate that its function is independent of AP-1, the transcriptional trigger of BMP signal transduction in leading edge cells. pMAD (phosphorylated Mothers Against Dpp) activity data show the mmy gene product to be a new type of epidermal BMP regulator - one which transforms a BMP ligand from a long- to a short-range signal. mmy codes for the single UDP-N-acetylglucosamine pyrophosphorylase in Drosophila, and its requirement for attenuating epidermal BMP signaling during dorsal closure points to a new role for glycosylation in defining a highly restricted BMP activity field in the fly. These findings add a new dimension to our understanding of mechanisms modulating the BMP signaling gradient.


Sujet(s)
Protéines de Drosophila/métabolisme , Drosophila melanogaster/enzymologie , Régulation de l'expression des gènes au cours du développement , Nucleotidyltransferases/métabolisme , Séquence d'acides aminés , Animaux , Protéines morphogénétiques osseuses/génétique , Protéines morphogénétiques osseuses/métabolisme , Protéines de Drosophila/génétique , Drosophila melanogaster/embryologie , Drosophila melanogaster/génétique , Embryon non mammalien/métabolisme , Développement embryonnaire , Activation enzymatique , Épiderme/métabolisme , Épiderme/anatomopathologie , Glycosylation , Système de signalisation des MAP kinases , Données de séquences moléculaires , Nucleotidyltransferases/génétique , Organismes génétiquement modifiés/embryologie , Organismes génétiquement modifiés/génétique , Organismes génétiquement modifiés/métabolisme , Structure tertiaire des protéines , Facteurs temps , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/métabolisme
17.
Cell Rep ; 3(5): 1355-61, 2013 May 30.
Article de Anglais | MEDLINE | ID: mdl-23707065

RÉSUMÉ

The presence of foreign DNA in the cytosol of mammalian cells elicits a potent antiviral interferon response. Recently, cytosolic DNA was proposed to induce the synthesis of cyclic GMP-AMP (cGAMP) upon binding to an enzyme called cGAMP synthase (cGAS). cGAMP activates an interferon response by binding to a downstream receptor called STING. Here, we identify natural variants of human STING (hSTING) that are poorly responsive to cGAMP yet, unexpectedly, are normally responsive to DNA and cGAS signaling. We explain this paradox by demonstrating that the cGAS product is actually a noncanonical cyclic dinucleotide, cyclic [G(2'-5')pA(3'-5')p], which contains a single 2'-5' phosphodiester bond. Cyclic [G(2'-5')pA(3'-5')p] potently activates diverse hSTING receptors and, therefore, may be a useful adjuvant or immunotherapeutic. Our results indicate that hSTING variants have evolved to distinguish conventional (3'-5') cyclic dinucleotides, known to be produced mainly by bacteria, from the noncanonical cyclic dinucleotide produced by mammalian cGAS.


Sujet(s)
Protéines membranaires/métabolisme , Nucléotides cycliques/métabolisme , Nucleotidyltransferases/métabolisme , Oligonucléotides/métabolisme , Animaux , Séquence nucléotidique , Lignée cellulaire , Cellules HEK293 , Humains , Immunité innée , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/génétique , Souris , Données de séquences moléculaires , Nucléotides cycliques/composition chimique , Oligonucléotides/composition chimique , Liaison aux protéines , Interférence par ARN , Petit ARN interférent/métabolisme
18.
Nature ; 478(7370): 515-8, 2011 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-21947006

RÉSUMÉ

The innate immune system detects infection by using germline-encoded receptors that are specific for conserved microbial molecules. The recognition of microbial ligands leads to the production of cytokines, such as type I interferons (IFNs), that are essential for successful pathogen elimination. Cytosolic detection of pathogen-derived DNA is one major mechanism of inducing IFN production, and this process requires signalling through TANK binding kinase 1 (TBK1) and its downstream transcription factor, IFN-regulatory factor 3 (IRF3). In addition, a transmembrane protein called STING (stimulator of IFN genes; also known as MITA, ERIS, MPYS and TMEM173) functions as an essential signalling adaptor, linking the cytosolic detection of DNA to the TBK1-IRF3 signalling axis. Recently, unique nucleic acids called cyclic dinucleotides, which function as conserved signalling molecules in bacteria, have also been shown to induce a STING-dependent type I IFN response. However, a mammalian sensor of cyclic dinucleotides has not been identified. Here we report evidence that STING itself is an innate immune sensor of cyclic dinucleotides. We demonstrate that STING binds directly to radiolabelled cyclic diguanylate monophosphate (c-di-GMP), and we show that unlabelled cyclic dinucleotides, but not other nucleotides or nucleic acids, compete with c-di-GMP for binding to STING. Furthermore, we identify mutations in STING that selectively affect the response to cyclic dinucleotides without affecting the response to DNA. Thus, STING seems to function as a direct sensor of cyclic dinucleotides, in addition to its established role as a signalling adaptor in the IFN response to cytosolic DNA. Cyclic dinucleotides have shown promise as novel vaccine adjuvants and immunotherapeutics, and our results provide insight into the mechanism by which cyclic dinucleotides are sensed by the innate immune system.


Sujet(s)
GMP cyclique/analogues et dérivés , Immunité innée/immunologie , Protéines membranaires/immunologie , Protéines membranaires/métabolisme , Adjuvants immunologiques , Séquence d'acides aminés , Animaux , GMP cyclique/immunologie , ADN/immunologie , Cellules HEK293 , Humains , Interférons/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Protéines membranaires/génétique , Souris , Données de séquences moléculaires
19.
Immunity ; 34(6): 854-65, 2011 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-21703540

RÉSUMÉ

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) proteins regulate innate immunity. Although the positive regulatory impact of NLRs is clear, their inhibitory roles are not well defined. We showed that Nlrx1(-/-) mice exhibited increased expression of antiviral signaling molecules IFN-ß, STAT2, OAS1, and IL-6 after influenza virus infection. Consistent with increased inflammation, Nlrx1(-/-) mice exhibited marked morbidity and histopathology. Infection of these mice with an influenza strain that carries a mutated NS-1 protein, which normally prevents IFN induction by interaction with RNA and the intracellular RNA sensor RIG-I, further exacerbated IL-6 and type I IFN signaling. NLRX1 also weakened cytokine responses to the 2009 H1N1 pandemic influenza virus in human cells. Mechanistically, Nlrx1 deletion led to constitutive interaction of MAVS and RIG-I. Additionally, an inhibitory function is identified for NLRX1 during LPS activation of macrophages where the MAVS-RIG-I pathway was not involved. NLRX1 interacts with TRAF6 and inhibits NF-κB activation. Thus, NLRX1 functions as a checkpoint of overzealous inflammation.


Sujet(s)
Sous-type H1N1 du virus de la grippe A/immunologie , Protéines mitochondriales/immunologie , Infections à Orthomyxoviridae/immunologie , Transduction du signal , Protéines adaptatrices de la transduction du signal/immunologie , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Cellules cultivées , Interféron bêta/biosynthèse , Interféron bêta/immunologie , Interleukine-6/biosynthèse , Interleukine-6/immunologie , Macrophages/immunologie , Protéines membranaires/immunologie , Protéines membranaires/métabolisme , Souris , Souris knockout , Protéines mitochondriales/déficit , Facteur de transcription NF-kappa B/immunologie , Facteur de transcription NF-kappa B/métabolisme , Protéines de tissu nerveux/immunologie , Protéines de tissu nerveux/métabolisme , Récepteurs de surface cellulaire , Facteur-6 associé aux récepteurs de TNF/immunologie , Facteur-6 associé aux récepteurs de TNF/métabolisme
20.
Infect Immun ; 79(2): 688-94, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-21098106

RÉSUMÉ

Type I interferons (IFNs) are central regulators of the innate and adaptive immune responses to viral and bacterial infections. Type I IFNs are induced upon cytosolic detection of microbial nucleic acids, including DNA, RNA, and the bacterial second messenger cyclic-di-GMP (c-di-GMP). In addition, a recent study demonstrated that the intracellular bacterial pathogen Listeria monocytogenes stimulates a type I IFN response due to cytosolic detection of bacterially secreted c-di-AMP. The transmembrane signaling adaptor Sting (Tmem173, Mita, Mpys, Eris) has recently been implicated in the induction of type I IFNs in response to cytosolic DNA and/or RNA. However, the role of Sting in response to purified cyclic dinucleotides or during in vivo L. monocytogenes infection has not been addressed. In order to identify genes important in the innate immune response, we have been conducting a forward genetic mutagenesis screen in C57BL/6 mice using the mutagen N-ethyl-N-nitrosourea (ENU). Here we describe a novel mutant mouse strain, Goldenticket (Gt), that fails to produce type I IFNs upon L. monocytogenes infection. By genetic mapping and complementation experiments, we found that Gt mice harbor a single nucleotide variant (T596A) of Sting that functions as a null allele and fails to produce detectable protein. Analysis of macrophages isolated from Gt mice revealed that Sting is absolutely required for the type I interferon response to both c-di-GMP and c-di-AMP. Additionally, Sting is required for the response to c-di-GMP and L. monocytogenes in vivo. Our results provide new functions for Sting in the innate interferon response to pathogens.


Sujet(s)
GMP cyclique/analogues et dérivés , Dinucléoside phosphates/métabolisme , Interféron de type I/métabolisme , Listeria monocytogenes/immunologie , Infections à Listeria/immunologie , Protéines membranaires/physiologie , Allèles , Animaux , Lignée cellulaire , GMP cyclique/métabolisme , 1-Éthyl-1-nitroso-urée/toxicité , Femelle , Régulation de l'expression des gènes/physiologie , Test de complémentation , Humains , Interféron de type I/génétique , Infections à Listeria/métabolisme , Macrophages/microbiologie , Mâle , Protéines membranaires/génétique , Souris , Souches mutantes de souris , Mutation , Polymorphisme de nucléotide simple
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE