972 resultados para TOLL-LIKE RECEPTORS
Resumo:
Summary The NACHT, LRR and PYD domains containing protein (NALP3) inflammasome is a key regulator of interleukin-1beta (IL-1beta) secretion. As there is strong evidence for a pro-inflammatory role of IL-1beta in rheumatoid arthritis (RA) and in murine models of arthritis, we explored the expression of the different components of the NALP3 inflammasome as well as other nucleotide oligomerization domain (NOD)-like receptors (NLRs) in synovium obtained from patients with RA. The expression of NLRs was also studied in fibroblast lines derived from joint tissue. By immunohistology, NALP3 and apoptosis-associated speck-like protein containing a CARD domain (ASC) were expressed in myeloid and endothelial cells and B cells. T cells expressed ASC but lacked NALP3. In synovial fibroblast lines, NALP3 expression was not detected at the RNA and protein levels and stimulation with known NALP3 agonists failed to induce IL-1beta secretion. Interestingly, we were unable to distinguish RA from osteoarthritis synovial samples on the basis of their basal level of RNA expression of known NLR proteins, though RA samples contained higher levels of caspase-1 assayed by enzyme-linked immunsorbent assay. These results indicate that myeloid and endothelial cells are the principal sources of inflammasome-mediated IL-1beta production in the synovium, and that synovial fibroblasts are unable to activate caspase-1 because they lack NALP3. The NALP3 inflammasome activity does not account for the difference in level of inflammation between RA and osteoarthritis.
NLRC5 deficiency selectively impairs MHC class I- dependent lymphocyte killing by cytotoxic T cells.
Resumo:
Nucleotide-binding oligomerization domain-like receptors (NLRs) are intracellular proteins involved in innate-driven inflammatory responses. The function of the family member NLR caspase recruitment domain containing protein 5 (NLRC5) remains a matter of debate, particularly with respect to NF-κB activation, type I IFN, and MHC I expression. To address the role of NLRC5, we generated Nlrc5-deficient mice (Nlrc5(Δ/Δ)). In this article we show that these animals exhibit slightly decreased CD8(+) T cell percentages, a phenotype compatible with deregulated MHC I expression. Of interest, NLRC5 ablation only mildly affected MHC I expression on APCs and, accordingly, Nlrc5(Δ/Δ) macrophages efficiently primed CD8(+) T cells. In contrast, NLRC5 deficiency dramatically impaired basal expression of MHC I in T, NKT, and NK lymphocytes. NLRC5 was sufficient to induce MHC I expression in a human lymphoid cell line, requiring both caspase recruitment and LRR domains. Moreover, endogenous NLRC5 localized to the nucleus and occupied the proximal promoter region of H-2 genes. Consistent with downregulated MHC I expression, the elimination of Nlrc5(Δ/Δ) lymphocytes by cytotoxic T cells was markedly reduced and, in addition, we observed low NLRC5 expression in several murine and human lymphoid-derived tumor cell lines. Hence, loss of NLRC5 expression represents an advantage for evading CD8(+) T cell-mediated elimination by downmodulation of MHC I levels-a mechanism that may be exploited by transformed cells. Our data show that NLRC5 acts as a key transcriptional regulator of MHC I in lymphocytes and support an essential role for NLRs in directing not only innate but also adaptive immune responses.
Resumo:
SUMMARY Nuclear factor kappa B (NF-κB) transcription factors control many aspects of cell fate through induction of inflammatory, immune or survival molecules. We have identified two novel proteins, named receptor interacting protein (RIP)-4 and caspase recruitment domain (CARD) adaptor inducing interferon-β (Cardif), which activate NF-κB. Further, we have found that Cardif plays a prominent antiviral function. Antiviral innate immunity is mounted upon recognition by the host of virally associated structures like double-stranded (ds) RNA, which constitutes a viral replication product of many viruses within infected cells. dsRNA, depending on its subcellular localization, can be sensed by two separate arms of host defense. Firstly, Toll-like receptor (TLR)-3, a member of the type I transmembrane TLR family, recognizes endosomally-located dsRNA. Secondly, cytoplasmic dsRNA is detected by the recently identified RNA helicase retinoic acid inducible gene I (RIG-I). Triggering of TLR3- and RIG-I-dependent pathways results in the activation of the transcription factors NF-κB and Interferon regulatory factor (IRF)-3, which cooperatively transduce antiviral immune responses. We have demonstrated that RIP1, a kinase previously shown to be required for TNF signaling, transmits TLR3-dependent NF-κB activation. Further we have identified and characterized Cardif as an essential adaptor transmitting RIG-I-mediated antiviral responses, including activation of NF-κB and IRF3. In addition, we showed that Cardif is cleaved and inactivated by a serine protease of hepatitis C virus, and therefore may represent an attractive target for this virus to escape innate immune responses. RESUME Les facteurs de transcription "nuclear factor kappa B" (NF-κB) contrôlent divers aspects du devenir cellulaire à travers l'induction de molécules inflammatoires, immunitaires ou de survie. Nous avons identifié deux nouvelles protéines, nommées "receptor interacting protein" (RIP)-4 et "caspase recruitment domain (CARD) adaptor inducing interferon-β" (Cardif), qui activent NF-κB. En outre, nous avons trouvé que Cardif joue un rôle antiviral crucial. L'immunité innée antivirale s'établit au moment de la reconnaissance par l'hôte de structures virales, comme l'ARN double brin, qui constitue un produit de réplication de beaucoup de virus à l'intérieur de cellules infectées. L'ARN double brin, dépendant de sa localisation subcellulaire, peut être détecté par deux branches de défense distinctes. Premièrement, le récepteur transmembranaire "Toll-like" (TLR), TLR3, reconnaît l'ARN double brin lorsque localisé dans les endosomes. Deuxièmement, l'ARN double brin cytoplasmique est reconnu par l'ARN hélicase récemment décrite "retinoic acid inducible gene I" (RIG-I). Le déclenchement de voies dépendantes de TLR3 et RIG-I active les facteurs de transcription NF-κB et IRF3, qui coopèrent afin de transduire des réponses immunitaires antivirales. Nous avons démontré que RIP1, une kinase décrite précédemment dans le signalement du TNF, transmet l'activation de NF-κB dépendante de TLR3. De plus, nous avons identifié et caractérisé Cardif comme un adapteur essentiel transmettant les réponses antivirales médiées par RIG-I, qui incluent l'activation de NF-κB et IRF3. De surcroît, Cardif est clivé et inactivé par une sérine protéase du virus de l'hépatite C, et ainsi pourrait représenter une cible attractive pour ce virus afin d'échapper aux réponses immunitaires innées.
Resumo:
Acute infection with the hepatitis C virus (HCV) induces a wide range of innate and adaptive immune responses. A total of 20-50% of acutely HCV-infected individuals permanently control the virus, referred to as 'spontaneous hepatitis C clearance', while the infection progresses to chronic hepatitis C in the majority of cases. Numerous studies have examined host genetic determinants of hepatitis C infection outcome and revealed the influence of genetic polymorphisms of human leukocyte antigens, killer immunoglobulin-like receptors, chemokines, interleukins and interferon-stimulated genes on spontaneous hepatitis C clearance. However, most genetic associations were not confirmed in independent cohorts, revealed opposing results in diverse populations or were limited by varying definitions of hepatitis C outcomes or small sample size. Coordinated efforts are needed in the search for key genetic determinants of spontaneous hepatitis C clearance that include well-conducted candidate genetic and genome-wide association studies, direct sequencing and follow-up functional studies.
Resumo:
The outcome of a viral infection depends on the interplay between the host's capacity to trigger potent antiviral responses and viral mechanisms that counteract them. Although Toll-like receptor (TLR)-3, which recognizes virally derived double-stranded (ds) RNA, transmits downstream antiviral signaling through the TIR adaptor Trif (TICAM-1), viral RNA-sensing RIG-like helicases (RLHs) use the mitochondrial-bound CARD protein Cardif (IPS-1/MAVS/VISA). The importance of these two antiviral signaling pathways is reflected by the fact that both adaptors are inhibited through specific cleavage triggered by the hepatitis C virus serine protease NS3-4A. Here, we show that inactivation can also occur through cellular caspases activated by various pro-apoptotic signals. Upon caspase-dependent cleavage both adaptors loose their capacity to activate the transcription factors interferon regulatory factors (IRF) and NF-kappaB. Importantly, poliovirus infection triggers a caspase-dependent cleavage of Cardif, suggesting that some viruses may activate caspases not only as a mean to facilitate shedding and replication, but also to impair antiviral responses
Resumo:
More than half of invasive bacterial infections are Gram-positive in origin. This class of bacteria has neither endotoxins nor an outer membrane, yet it generates some of the most powerful inflammatory responses known in medicine. Some recent seminal studies go a long way toward settling the controversies that surround the process by which Gram-positive bacterial surfaces trigger the human immune system. Although the components of the cell wall are now chemically defined in exquisite detail and the interaction with the toll-like receptor 2 pathway has been discovered, it is only very recently that definitive studies combining these advanced biochemical and cell biological tools have been carried out. It is these breakthrough studies that have finally confirmed the paradigm of innate sensors for Gram-positive bacteria.
Resumo:
OBJECTIVES: To evaluate the influence of genetic polymorphisms on the susceptibility to Candida colonization and intra-abdominal candidiasis, a blood culture-negative life-threatening infection in high-risk surgical ICU patients. DESIGN: Prospective observational cohort study. SETTING: Surgical ICUs from two University hospitals of the Fungal Infection Network of Switzerland. PATIENTS: Eighty-nine patients at high risk for intra-abdominal candidiasis (68 with recurrent gastrointestinal perforation and 21 with acute necrotizing pancreatitis). MEASUREMENTS AND MAIN RESULTS: Eighteen single-nucleotide polymorphisms in 16 genes previously associated with development of fungal infections were analyzed from patient's DNA by using an Illumina Veracode genotyping platform. Candida colonization was defined by recovery of Candida species from at least one nonsterile site by twice weekly monitoring of cultures from oropharynx, stools, urine, skin, and/or respiratory tract. A corrected colonization index greater than or equal to 0.4 defined "heavy" colonization. Intra-abdominal candidiasis was defined by the presence of clinical symptoms and signs of peritonitis or intra-abdominal abscess and isolation of Candida species either in pure or mixed culture from intraoperatively collected abdominal samples. Single-nucleotide polymorphisms in three innate immune genes were associated with development of a Candida corrected colonization index greater than or equal to 0.4 (Toll-like receptor rs4986790, hazard ratio = 3.39; 95% CI, 1.45-7.93; p = 0.005) or occurrence of intra-abdominal candidiasis (tumor necrosis factor-α rs1800629, hazard ratio = 4.31; 95% CI, 1.85-10.1; p= 0.0007; β-defensin 1 rs1800972, hazard ratio = 3.21; 95% CI, 1.36-7.59; p = 0.008). CONCLUSION: We report a strong association between the promoter rs1800629 single-nucleotide polymorphism in tumor necrosis factor-α and an increased susceptibility to intra-abdominal candidiasis in a homogenous prospective cohort of high-risk surgical ICU patients. This finding highlights the relevance of the tumor necrosis factor-α functional polymorphism in immune response to fungal pathogens. Immunogenetic profiling in patients at clinical high risk followed by targeted antifungal interventions may improve the prevention or preemptive management of this life-threatening infection.
Resumo:
BACKGROUND & AIMS: Priming of T cells by dendritic cells (DCs) in the intestinal mucosa and associated lymphoid tissues helps maintain mucosal tolerance but also contributes to the development of chronic intestinal inflammation. Chemokines regulate the intestinal immune response and can contribute to pathogenesis of inflammatory bowel diseases. We investigated the role of the chemokine CCL17, which is expressed by conventional DCs in the intestine and is up-regulated during colitis. METHODS: Colitis was induced by administration of dextran sodium sulfate (DSS) to mice or transfer of T cells to lymphopenic mice. Colitis activity was monitored by body weight assessment, histologic scoring, and cytokine profile analysis. The direct effects of CCL17 on DCs and the indirect effects on differentiation of T helper (Th) cells were determined in vitro and ex vivo. RESULTS: Mice that lacked CCL17 (Ccl17(E/E) mice) were protected from induction of severe colitis by DSS or T-cell transfer. Colonic mucosa and mesenteric lymph nodes from Ccl17-deficient mice produced lower levels of proinflammatory cytokines. The population of Foxp3(+) regulatory T cells (Tregs) was expanded in Ccl17(E/E) mice and required for long-term protection from colitis. CCR4 expression by transferred T cells was not required for induction of colitis, but CCR4 expression by the recipients was required. CCL17 promoted Toll-like receptor-induced secretion of interleukin-12 and interleukin-23 by DCs in an autocrine manner, promoted differentiation of Th1 and Th17 cells, and reduced induction of Foxp3(+) Treg cells. CONCLUSIONS: The chemokine CCL17 is required for induction of intestinal inflammation in mice. CCL17 has an autocrine effect on DCs that promotes production of inflammatory cytokines and activation of Th1 and Th17 cells and reduces expansion of Treg cells.
Resumo:
A current paradigm proposes that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3 inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Toll-like-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome activation and pro-inflammatory cytokine levels.
Resumo:
OBJECTIVE: Monosodium urate monohydrate (MSU) crystal-induced interleukin-1β (IL-1β) secretion is a critical factor in the pathogenesis of gout. However, without costimulation by a proIL-1β-inducing factor, MSU crystals alone are insufficient to induce IL-1β secretion. The responsible costimulatory factors that act as a priming endogenous signal in vivo are not yet known. We undertook this study to analyze the costimulatory properties of myeloid-related protein 8 (MRP-8) and MRP-14 (endogenous Toll-like receptor 4 [TLR-4] agonists) in MSU crystal-induced IL-1β secretion and their relevance in gout. METHODS: MRP-8/MRP-14 was measured in paired serum and synovial fluid samples by enzyme-linked immunosorbent assay (ELISA) and localized in synovial tissue from gout patients by immunohistochemistry. Serum levels were correlated with disease activity, and MSU crystal-induced release of MRPs from human phagocytes was measured. Costimulatory effects of MRP-8 and MRP-14 on MSU crystal-induced IL-1β secretion from phagocytes were analyzed in vitro by ELISA, Western blotting, and polymerase chain reaction. The impact of MRP was tested in vivo in a murine MSU crystal-induced peritonitis model. RESULTS: MRP-8/MRP-14 levels were elevated in the synovium, tophi, and serum of patients with gout and correlated with disease activity. MRP-8/MRP-14 was released by MSU crystal-activated phagocytes and increased MSU crystal-induced IL-1β secretion in a TLR-4-dependent manner. Targeted deletion of MRP-14 in mice led to a moderately reduced response of MSU crystal-induced inflammation in vivo. CONCLUSION: MRP-8 and MRP-14, which are highly expressed in gout, are enhancers of MSU crystal-induced IL-1β secretion in vitro and in vivo. These endogenous TLR-4 ligands released by activated phagocytes contribute to the maintenance of inflammation in gout.
Resumo:
A genome-wide screen for large structural variants showed that a copy number variant (CNV) in the region encoding killer cell immunoglobulin-like receptors (KIR) associates with HIV-1 control as measured by plasma viral load at set point in individuals of European ancestry. This CNV encompasses the KIR3DL1-KIR3DS1 locus, encoding receptors that interact with specific HLA-Bw4 molecules to regulate the activation of lymphocyte subsets including natural killer (NK) cells. We quantified the number of copies of KIR3DS1 and KIR3DL1 in a large HIV-1 positive cohort, and showed that an increase in KIR3DS1 count associates with a lower viral set point if its putative ligand is present (p = 0.00028), as does an increase in KIR3DL1 count in the presence of KIR3DS1 and appropriate ligands for both receptors (p = 0.0015). We further provide functional data that demonstrate that NK cells from individuals with multiple copies of KIR3DL1, in the presence of KIR3DS1 and the appropriate ligands, inhibit HIV-1 replication more robustly, and associated with a significant expansion in the frequency of KIR3DS1+, but not KIR3DL1+, NK cells in their peripheral blood. Our results suggest that the relative amounts of these activating and inhibitory KIR play a role in regulating the peripheral expansion of highly antiviral KIR3DS1+ NK cells, which may determine differences in HIV-1 control following infection.
Resumo:
SummaryMulticellular organisms have evolved an immune system in order to cope with the constant threats they are facing. Foreign pathogens or endogenous danger signals released by injured or dying host cells can be readily detected through a set of germline- encoded pattern-recognition receptors. The NOD-like receptors are a cytoplasmic family of pattern-recognition receptors that have recently attracted considerable attention due to their ability to form inflammasomes, which are molecular complexes responsible for the activation of caspase-1 and the subsequent processing of the pro¬inflammatory cytokines IL-IB and 11-18 into their mature, bioactive form.In this study, we describe a novel pro-inflammatory signaling pathway, whereby the endoplasmic reticulum promotes inflammation through activation of the NLRP3 inflammasome. This was shown to be independent of the classical endoplasmic reticulum stress response pathway constituted by the effectors IREla, PERK and ATF6a. In keeping with other known NLRP3 activators, generation of reactive oxygen species and potassium efflux were required. We also provide evidence that calcium signaling is critical to this pathway, and possibly integrates signaling triggered by various NLRP3 inflammasome activators. Moreover, the mitochondrial channel VDAC1 was instrumental in mediating this response. We thus propose that the endoplasmic reticulum acts as an integrator of stress and is able to activate the mitochondria in a calcium-dependent manner in order to promote NLRP3 inflammasome activation in response to a wide range of activators.Given the role played by inflammation in the pathogenesis of atherosclerosis, we decided to investigate a possible role for the NLRP3 inflammasome in the progression of the disease. Using an ApoE mouse model, we find that deficiency in the NLRP3 inflammasome components NLRP3, ASC or Caspase-1 does not impair atherosclerosis progression, nor does it impact plaque stability. While previous studies have clearly shown a role for the interleukin-1 family of ligands in atherosclerosis, our results suggest that its contribution might be more complex than previously appreciated, and further research is thus warranted in this field.RésuméLes organismes multicellulaires ont développé un système immunitaire pour faire face aux menaces qui les entourent. Des pathogènes étrangers ou des signaux de danger relâchés par des cellules de l'hôte en détresse peuvent être rapidement détectés via un assemblage de récepteurs spécifiques qui sont présents dès la naissance. Certains membres de la famille de récepteurs NOD ont récemment attiré beaucoup d'attention au vu de leur capacité à former des inflammasomes, complexes moléculaires responsables de l'activation de la easpase-1 et de la maturation des cytokines pro-inflammatoires IL- 1β et IL-18 en leur forme bioactive.Dans cette étude, nous décrivons une nouvelle voie de signalisation pro-inflammatoire, par laquelle le réticulum endoplasmique induit l'inflammation via l'activation de l'inflammasome NLRP3. Cette voie est indépendante de la voie classique de réponse au stress du réticulum endoplasmique, qui comprend les effecteurs IRE1, PERK et ATF6. Comme pour d'autres activateurs de NLRP3, la génération de radicaux libres d'oxygène ainsi que Γ efflux de potassium sont requis. Nous montrons également que le calcium joue un rôle critique dans cette voie, et intègre possiblement la signalisation provoquée par divers activateurs de l'inflammasome NLRP3. De plus, le canal mitochondrial VDAC1 est essentiel dans cette réponse. Nous proposons donc que le réticulum endoplasmique agit comme un intégrateur de stress, activant la mitochondrie d'une façon calcium-dépendante pour promouvoir l'activation de l'inflammasome NLRP3 en réponse à divers activateurs.Au vu du rôle joué par l'inflammation dans la pathogenèse de l'athérosclérose, nous avons étudié un possible rôle pour l'inflammasome NLRP3 dans la progression de la maladie. Dans un modèle de souris ApoE, l'absence des composants de l'inflammasome NLRP3 que sont NLRP3, ASC et Caspase-1 n'influence pas la progression des plaques ni leur stabilité. Alors que d'autres études ont démontré un rôle pour les membres de la famille de l'interleukine-1 dans l'athérosclérose, nos résultats suggèrent que leur contribution pourrait être plus complexe que précédemment apprécié, et d'autres recherches dans ce domaine sont donc nécessaires.
Resumo:
In AKI, dying renal cells release intracellular molecules that stimulate immune cells to secrete proinflammatory cytokines, which trigger leukocyte recruitment and renal inflammation. Whether the release of histones, specifically, from dying cells contributes to the inflammation of AKI is unknown. In this study, we found that dying tubular epithelial cells released histones into the extracellular space, which directly interacted with Toll-like receptor (TLR)-2 (TLR2) and TLR4 to induce MyD88, NF-κB, and mitogen activated protein kinase signaling. Extracellular histones also had directly toxic effects on renal endothelial cells and tubular epithelial cells in vitro. In addition, direct injection of histones into the renal arteries of mice demonstrated that histones induce leukocyte recruitment, microvascular vascular leakage, renal inflammation, and structural features of AKI in a TLR2/TLR4-dependent manner. Antihistone IgG, which neutralizes the immunostimulatory effects of histones, suppressed intrarenal inflammation, neutrophil infiltration, and tubular cell necrosis and improved excretory renal function. In summary, the release of histones from dying cells aggravates AKI via both its direct toxicity to renal cells and its proinflammatory effects. Because the induction of proinflammatory cytokines in dendritic cells requires TLR2 and TLR4, these results support the concept that renal damage triggers an innate immune response, which contributes to the pathogenesis of AKI.
Resumo:
During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.
Resumo:
BACKGROUND: Macrophage migration inhibitory factor (MIF) has emerged as a pivotal mediator of innate immunity and has been shown to be an important effector molecule in severe sepsis. Melioidosis, caused by Burkholderia pseudomallei, is an important cause of community-acquired sepsis in Southeast-Asia. We aimed to characterize the expression and function of MIF in melioidosis. METHODOLOGY AND PRINCIPAL FINDINGS: MIF expression was determined in leukocytes and plasma from 34 melioidosis patients and 32 controls, and in mice infected with B. pseudomallei. MIF function was investigated in experimental murine melioidosis using anti-MIF antibodies and recombinant MIF. Patients demonstrated markedly increased MIF mRNA leukocyte and MIF plasma concentrations. Elevated MIF concentrations were associated with mortality. Mice inoculated intranasally with B. pseudomallei displayed a robust increase in pulmonary and systemic MIF expression. Anti-MIF treated mice showed lower bacterial loads in their lungs upon infection with a low inoculum. Conversely, mice treated with recombinant MIF displayed a modestly impaired clearance of B. pseudomallei. MIF exerted no direct effects on bacterial outgrowth or phagocytosis of B. pseudomallei. CONCLUSIONS: MIF concentrations are markedly elevated during clinical melioidosis and correlate with patients' outcomes. In experimental melioidosis MIF impaired antibacterial defense.