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










Publication year range
1.
J Immunol ; 212(12): 1904-1912, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38668728

ABSTRACT

NK cells have been shown to exhibit inflammatory and immunoregulatory functions in a variety of healthy and diseased settings. In the context of chronic viral infection and cancer, distinct NK cell populations that inhibit adaptive immune responses have been observed. To understand how these cells arise and further characterize their immunosuppressive role, we examined in vitro conditions that could polarize human NK cells into an inhibitory subset. TGF-ß1 has been shown to induce regulatory T cells in vitro and in vivo; we therefore investigated if TGF-ß1 could also induce immunosuppressive NK-like cells. First, we found that TGF-ß1/IL-15, but not IL-15 alone, induced CD103+CD49a+ NK-like cells from peripheral blood NK cells, which expressed markers previously associated with inhibitory CD56+ innate lymphoid cells, including high expression of GITR and CD101. Moreover, supernatant from ascites collected from patients with ovarian carcinoma also induced CD103+CD49a+ NK-like cells in vitro in a TGF-ß-dependent manner. Interestingly, TGF-ß1/IL-15-induced CD103+CD56+ NK-like cells suppressed autologous CD4+ T cells in vitro by reducing absolute number, proliferation, and expression of activation marker CD25. Collectively, these findings provide new insight into how NK cells may acquire an inhibitory phenotype in TGF-ß1-rich environments.


Subject(s)
Interleukin-15 , Killer Cells, Natural , Transforming Growth Factor beta1 , Humans , Killer Cells, Natural/immunology , Interleukin-15/immunology , Interleukin-15/metabolism , Transforming Growth Factor beta1/metabolism , Female , Antigens, CD/metabolism , Antigens, CD/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Integrin alpha Chains/metabolism , Integrin alpha Chains/immunology , CD56 Antigen/metabolism , Cells, Cultured , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Activation/immunology
2.
Cancer Immunol Immunother ; 72(7): 2375-2392, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36943460

ABSTRACT

Immunotherapeutic strategies aimed at enhancing tumor cell killing by tumor-specific T cells hold great potential for reducing tumor burden and prolonging survival of cancer patients. Although many potential tumor antigens have been described, identifying relevant targets when designing anti-cancer vaccines or targeted cell therapies remains a challenge. To identify novel, potentially immunogenic candidate tumor antigens, we performed integrated tumor transcriptomic, seromic, and proteomic analyses of high grade serous ovarian cancer (HGSC) patient tumor samples. We identified tumor neo-antigens and over-expressed antigens using whole exome and RNA sequencing and examined these in relation to patient-matched auto-antibody repertoires. Focusing on MHC class I epitopes recognized by CD8+ T cells, HLA-binding epitopes were identified or predicted from the highly expressed, mutated, or auto-antibody target antigen, or MHC-associated peptides (MAPs). Recognition of candidate antigenic peptides was assessed within the tumor-infiltrating T lymphocyte (TIL) population expanded from each patient. Known tumor-associated antigens (TAA) and cancer/testis antigens (CTA) were commonly found in the auto-antibody and MAP repertoires and CD8+ TILs recognizing epitopes from these antigens were detected, although neither expression level nor the presence of auto-antibodies correlated with TIL recognition. Auto-antibodies against tumor-mutated antigens were found in most patients, however, no TIL recognition of the highest predicted affinity neo-epitopes was detected. Using high expression level, auto-antibody recognition, and epitope prediction algorithms, we identified epitopes in 5 novel antigens (MOB1A, SOCS3, TUBB, PRKAR1A, CCDC6) recognized by HGSC patient TILs. Furthermore, selection of epitopes from the MAP repertoire identified 5 additional targets commonly recognized by multiple patient TILs. We find that the repertoire of TIL specificities includes recognition of highly expressed and immunogenic self-antigens that are processed and presented by tumors. These results indicate an ongoing autoimmune response against a range of self-antigens targeted by HGSC TILs.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Ovarian Neoplasms , Male , Humans , Female , Epitopes/metabolism , CD8-Positive T-Lymphocytes , Proteomics , Multiomics , Antigens, Neoplasm , Peptides , Autoantigens , Epitopes, T-Lymphocyte
3.
Elife ; 92020 04 21.
Article in English | MEDLINE | ID: mdl-32314731

ABSTRACT

HLA-restricted T cell responses can induce antitumor effects in cancer patients. Previous human T cell research has largely focused on the few HLA alleles prevalent in a subset of ethnic groups. Here, using a panel of newly developed peptide-exchangeable peptide/HLA multimers and artificial antigen-presenting cells for 25 different class I alleles and greater than 800 peptides, we systematically and comprehensively mapped shared antigenic epitopes recognized by tumor-infiltrating T lymphocytes (TILs) from eight melanoma patients for all their class I alleles. We were able to determine the specificity, on average, of 12.2% of the TILs recognizing a mean of 3.1 shared antigen-derived epitopes across HLA-A, B, and C. Furthermore, we isolated a number of cognate T cell receptor genes with tumor reactivity. Our novel strategy allows for a more complete examination of the immune response and development of novel cancer immunotherapy not limited by HLA allele prevalence or tumor mutation burden.


The immune system is the body's way of defending itself, offering protection against diseases such as cancer. But to remove the cancer cells, the immune system must be able to identify them as different from the rest of the body. All cells break down proteins into shorter fragments, known as peptides, that are displayed on the cell surface by a protein called human leukocyte antigen, HLA for short. Cancer cells display distinctive peptides on their surface as they generate different proteins to those of healthy cells. Immune cells called T cells use these abnormal peptides to identify the cancer so that it can be destroyed. Sometimes T cells can lack the right equipment to detect abnormal peptides, allowing cancer cells to hide from the immune system. However, T cells can be trained through a treatment called immunotherapy, which provides T cells with new tools so that they can spot the peptides displayed by HLA on the previously 'hidden' cancer cells. There are many different forms of HLA, each of which can display different peptides. Current research in immunotherapy commonly targets only a subset of HLA forms, and not all cancer patients have these types. This means that immunotherapy research is only likely to be of most benefit to a limited number of patients. Immunotherapy could be made effective for more people if new cancer peptides that are displayed by the other 'under-represented' forms of HLA were identified. Murata, Nakatsugawa et al. have now used T cells that were taken from tumors in eight patients with melanoma, which is a type of skin cancer. A library of fluorescent HLA-peptides was generated ­ using a new, simplified methodology ­ with 25 forms of HLA that displayed over 800 peptides. T cells were then mixed with the library to identify which HLA-peptides they can target. As a result, Murata, Nakatsugawa et al. found the cancer targets of around 12% of the tumor-infiltrating T cells tested, including those from under-represented forms of HLA. Consequently, these findings could be used to develop new immunotherapies that can treat more patients.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Humans , Receptors, Antigen, T-Cell/immunology
4.
Cancer Immunol Res ; 8(4): 544-555, 2020 04.
Article in English | MEDLINE | ID: mdl-32047025

ABSTRACT

High-grade serous ovarian cancer (HGSC), the principal cause of death from gynecologic malignancies in the world, has not significantly benefited from advances in cancer immunotherapy. Although HGSC infiltration by lymphocytes correlates with superior survival, the nature of antigens that can elicit anti-HGSC immune responses is unknown. The goal of this study was to establish the global landscape of HGSC tumor-specific antigens (TSA) using a mass spectrometry pipeline that interrogated all reading frames of all genomic regions. In 23 HGSC tumors, we identified 103 TSAs. Classic TSA discovery approaches focusing only on mutated exonic sequences would have uncovered only three of these TSAs. Other mutated TSAs resulted from out-of-frame exonic translation (n = 2) or from noncoding sequences (n = 7). One group of TSAs (n = 91) derived from aberrantly expressed unmutated genomic sequences, which were not expressed in normal tissues. These aberrantly expressed TSAs (aeTSA) originated primarily from nonexonic sequences, in particular intronic (29%) and intergenic (22%) sequences. Their expression was regulated at the transcriptional level by variations in gene copy number and DNA methylation. Although mutated TSAs were unique to individual tumors, aeTSAs were shared by a large proportion of HGSCs. Taking into account the frequency of aeTSA expression and HLA allele frequencies, we calculated that, in Caucasians, the median number of aeTSAs per tumor would be five. We conclude that, in view of their number and the fact that they are shared by many tumors, aeTSAs may be the most attractive targets for HGSC immunotherapy.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Cystadenocarcinoma, Serous/pathology , Immunotherapy/methods , Mutation , Ovarian Neoplasms/pathology , Proteogenomics/methods , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism
5.
Sci Immunol ; 3(30)2018 12 14.
Article in English | MEDLINE | ID: mdl-30552102

ABSTRACT

Antigen recognition by T cells bearing αß T cell receptors (TCRs) is restricted by major histocompatibility complex (MHC). However, how antigens are recognized by T cells bearing γδ TCRs remains unclear. Although γδ T cells can recognize nonclassical MHC, it is generally thought that recognition of antigens is not MHC restricted. Here, we took advantage of an in vitro system to generate antigen-specific human T cells and show that melanoma-associated antigens, MART-1 and gp100, can be recognized by γδ T cells in an MHC-restricted fashion. Cloning and transferring of MART-1-specific γδ TCRs restored the specific recognition of the initial antigen MHC/peptide reactivity and conferred antigen-specific functional responses. A crystal structure of a MART-1-specific γδ TCR, together with MHC/peptide, revealed distinctive but similar docking properties to those previously reported for αß TCRs, recognizing MART-1 on HLA-A*0201. Our work shows that antigen-specific and MHC-restricted γδ T cells can be generated in vitro and that MART-1-specific γδ T cells can also be found and cloned from the naïve repertoire. These findings reveal that classical MHC-restricted human γδ TCRs exist in the periphery and have the potential to be used in developing of new TCR-based immunotherapeutic approaches.


Subject(s)
MART-1 Antigen/immunology , Melanoma/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Adult , Cells, Cultured , Crystallography, X-Ray , Humans , MART-1 Antigen/chemistry , Models, Molecular , Receptors, Antigen, T-Cell, gamma-delta/chemistry
7.
J Immunol ; 195(10): 4632-40, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26447227

ABSTRACT

TLR-induced maturation of dendritic cells (DCs) leads to the production of proinflammatory cytokines as well as the upregulation of various molecules involved in T cell activation. These are believed to be the critical events that account for the induction of the adaptive immune response. In this study, we have examined the role of miR-155 in DC function and the induction of immunity. Using a model in which the transfer of self-Ag-pulsed, TLR-matured DCs can induce a functional CD8 T cell response and autoimmunity, we find that DCs lacking miR-155 have an impaired ability to break immune tolerance. Importantly, transfer of self- Ag-pulsed DCs overexpressing miR-155 was sufficient to break tolerance in the absence of TLR stimuli. Although these unstimulated DCs induced T cell function in vivo, there was no evidence for the upregulation of costimulatory ligands or cytokine secretion. Further analysis showed that miR-155 influenced the level of the phosphatase SHIP1 in DCs and that the lack of SHIP1 in DCs was sufficient to break T cell tolerance in vivo, again in the absence of TLR-induced DC maturation. Our study demonstrates that the overexpression of miR-155 in DCs is a critical event that is alone sufficient to break self-tolerance and promote a CD8-mediated autoimmune response in vivo. This process is independent of the induction of conventional DC maturation markers, indicating that miR-155 regulation of SHIP represents a unique axis that regulates DC function in vivo.


Subject(s)
Dendritic Cells/metabolism , Immune Tolerance/immunology , MicroRNAs/biosynthesis , Phosphoric Monoester Hydrolases/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Immune Tolerance/genetics , Inositol Polyphosphate 5-Phosphatases , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Up-Regulation
8.
Macromol Biosci ; 14(11): 1528-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25130689

ABSTRACT

A versatile approach is proposed for the synthesis of novel immunoactive nanomaterials based on biocompatible poly(glycerol monomethacrylate) (PGMMA). Propargyl-terminated PGMMA is synthesized via atom transfer radical polymerization and then modified through the introduction of dangling acrylate groups, at controlled degree of functionalisation. Acrylates are then able to react quantitatively with thiols, such as immunoactive thiomannose, through Michael-type addition under mild conditions and at a physiologically acceptable pH. The terminal propargyl group can be modified independently with azide end-capping groups and it is utilized to graft the macromolecules to a fluorescent dye. The resulting mannose-linked PGMMAs confirm a safe cytotoxic profile and are able to stimulate cytokine production (TNFα), membrane protein expression (CD40), and cellular uptake in bone marrow derived dendritic cells. Cell stimulation is dependent on the mannose content and enhanced by serum proteins, suggesting a role for mannose-binding receptors and/or complement receptors in the cell membrane.


Subject(s)
Click Chemistry/methods , Glycerol/analogs & derivatives , Glycerol/chemical synthesis , Immunization , Nanostructures/chemistry , Polymethacrylic Acids/chemical synthesis , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , CD40 Antigens/metabolism , Cell Survival/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Fluorescent Dyes/metabolism , Glycerol/chemistry , Glycerol/pharmacology , Kinetics , Mice, Inbred C57BL , Molecular Weight , Polymerization , Polymethacrylic Acids/chemistry , Polymethacrylic Acids/pharmacology , Proton Magnetic Resonance Spectroscopy , Tumor Necrosis Factor-alpha/metabolism
9.
PLoS One ; 9(3): e92380, 2014.
Article in English | MEDLINE | ID: mdl-24647761

ABSTRACT

Vaccines for cancer immunotherapy are of interest but in general have not yet achieved the desired therapeutic efficacy in clinical trials. We present here a novel model to evaluate vaccine strategies by following tissue destruction in a transgenic model, where a defined antigen is expressed on pancreatic islets. We found that the transfer of syngeneic antigen-pulsed dendritic cells (DCs) resulted in autoimmune cytotoxic T-lymphocyte activation that was not observed following vaccinations that were based on peptides and adjuvants. Importantly, the induction of diabetes by DC transfer is dependent upon the maturation of DCs prior to transfer. Furthermore, diabetes induction only occurred if DCs were pulsed with the immunodominant epitope in addition to at least one other peptide, suggesting greater cytolytic activity upon engagement of multiple T-cell specificities. While the tumor environment undoubtedly will be more complex than healthy tissue, the insights gained through this model provide useful information on variables that can affect CD8-mediated tissue cytolysis in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Peptides/pharmacology , Animals , Cells, Cultured , Humans , Mice , T-Lymphocytes, Cytotoxic/immunology
10.
J Immunol ; 183(9): 5728-37, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812207

ABSTRACT

Inflammation following tissue damage promotes lymphocyte recruitment, tissue remodeling, and wound healing while maintaining self tolerance. Endogenous signals associated with tissue damage and cell death have been proposed to initiate and instruct immune responses following injury. In this study, we have examined the effects of elevated levels of a candidate endogenous danger signal, heat shock cognate protein 70 (hsc70), on stimulation of inflammation and autoimmunity following cell damage. We find that damage to pancreatic beta cells expressing additional cytosolic hsc70 leads to an increased incidence of diabetes in a transgenic mouse model. Steady-state levels of activated APC and T cell populations in the draining lymph node were enhanced, which further increased following streptozotocin-induced beta cell death. In addition, proinflammatory serum cytokines, and lymphocyte recruitment were increased in hsc70 transgenic mice. Islet Ag-specific T cells underwent a greater extent of proliferation in the lymph nodes of mice expressing hsc70 following beta cell damage, suggesting elevated Ag presentation following release of Ag in the presence of hsc70. These findings suggest that an elevated content of hsc70 in cells undergoing necrotic or apoptotic cell death can increase the extent of sterile inflammation and increase the susceptibility to autoimmunity.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , HSC70 Heat-Shock Proteins/biosynthesis , HSC70 Heat-Shock Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Animals , Autoimmune Diseases/epidemiology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cattle , Cell Death/genetics , Cell Death/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytosol/immunology , Cytosol/metabolism , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Incidence , Islets of Langerhans/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Rats
11.
Science ; 311(5769): 1927-32, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16574867

ABSTRACT

IRAK-4 is a protein kinase that is pivotal in mediating signals for innate immune responses. Here, we report that IRAK-4 signaling is also essential for eliciting adaptive immune responses. Thus, in the absence of IRAK-4, in vivo T cell responses were significantly impaired. Upon T cell receptor stimulation, IRAK-4 is recruited to T cell lipid rafts, where it induces downstream signals, including protein kinase C activation through the association with Zap70. This signaling pathway was found to be required for optimal activation of nuclear factor kappaB. Our findings suggest that T cells use this critical regulator of innate immunity for the development of acquired immunity, suggesting that IRAK-4 may be involved in direct signal cross talk between the two systems.


Subject(s)
Lymphocyte Activation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , T-Lymphocytes/immunology , Animals , Enzyme Activation , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases , Isoenzymes/metabolism , Membrane Microdomains/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , Receptors, Antigen, T-Cell/immunology , Signal Transduction , ZAP-70 Protein-Tyrosine Kinase/metabolism
12.
Nat Med ; 9(12): 1469-76, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625545

ABSTRACT

Pathogens or pathogen-associated molecular patterns can signal to cells of the innate immune system and trigger effective adaptive immunity. However, relatively little is known about how the innate immune system detects tissue injury or necrosis. Evidence suggests that the release of heat-shock proteins (HSPs) may provide adjuvant-like signals, but the ability of HSPs to promote activation or tolerance in vivo has not been addressed. In this study we show that Hsp70 promotes dendritic cell (DC) function and, together with antigen, triggers autoimmune disease in vivo.


Subject(s)
Antigen-Presenting Cells/immunology , HSP70 Heat-Shock Proteins/immunology , T-Lymphocytes/immunology , Animals , Autoimmunity , Base Sequence , CD40 Antigens/metabolism , DNA, Complementary/genetics , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Humans , Immune Tolerance , In Vitro Techniques , Interleukin-12/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/immunology , Signal Transduction
13.
J Immunol ; 170(8): 4031-5, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682231

ABSTRACT

IL-18 is an important cytokine for both innate and adaptive immunity. NK T cells and Th1 cells depend on IL-18 for their divergent functions. The IL-18R, IL-1R, and mammalian Toll-like receptors (TLRs) share homologous intracellular domains known as the TLR/IL-1R/plant R domain. Previously, we reported that IL-1R-associated kinase (IRAK)-4 plays a critical role in IL-1R and TLR signaling cascades and is essential for the innate immune response. Because TLR/IL-1R/plant R-containing receptors mediate signal transduction in a similar fashion, we investigated the role of IRAK-4 in IL-18R signaling. In this study, we show that IL-18-induced responses such as NK cell activity, Th1 IFN-gamma production, and Th1 cell proliferation are severely impaired in IRAK-4-deficient mice. IRAK-4(-/-) Th1 cells also do not exhibit NF-kappaB activation or IkappaB degradation in response to IL-18. Moreover, AP-1 activation which is triggered by c-Jun N-terminal kinase activation is also completely inhibited in IRAK-4(-/-) Th1 cells. These results suggest that IRAK-4 is an essential component of the IL-18 signaling cascade.


Subject(s)
Interleukin-18/physiology , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Receptors, Interleukin-1/metabolism , Th1 Cells/enzymology , Th1 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Interferon-gamma/biosynthesis , Interleukin-1 Receptor-Associated Kinases , Interleukin-18/metabolism , Interleukin-18 Receptor alpha Subunit , Killer Cells, Natural/metabolism , Mice , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/genetics , Receptors, Interleukin-18 , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/enzymology , Th2 Cells/immunology
14.
Nature ; 416(6882): 750-6, 2002 Apr 18.
Article in English | MEDLINE | ID: mdl-11923871

ABSTRACT

Toll-like receptors (TLRs), which recognize pathogen-associated molecular patterns, and members of the pro-inflammatory interleukin-1 receptor (IL-1R) family, share homologies in their cytoplasmic domains called Toll/IL-1R/plant R gene homology (TIR) domains. Intracellular signalling mechanisms mediated by TIRs are similar, with MyD88 (refs 5-8) and TRAF6 (refs 9, 10) having critical roles. Signal transduction between MyD88 and TRAF6 is known to involve the serine-threonine kinase IL-1 receptor-associated kinase 1 (IRAK-1) and two homologous proteins, IRAK-2 (ref. 12) and IRAK-M. However, the physiological functions of the IRAK molecules remain unclear, and gene-targeting studies have shown that IRAK-1 is only partially required for IL-1R and TLR signalling. Here we show by gene-targeting that IRAK-4, an IRAK molecule closely related to the Drosophila Pelle protein, is indispensable for the responses of animals and cultured cells to IL-1 and ligands that stimulate various TLRs. IRAK-4-deficient animals are completely resistant to a lethal dose of lipopolysaccharide (LPS). In addition, animals lacking IRAK-4 are severely impaired in their responses to viral and bacterial challenges. Our results indicate that IRAK-4 has an essential role in innate immunity.


Subject(s)
Drosophila Proteins , Membrane Glycoproteins/metabolism , Protein Kinases/deficiency , Protein Kinases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Gene Deletion , Immunity, Innate/immunology , Interferon-gamma/analysis , Interleukin-1/biosynthesis , Interleukin-1/pharmacology , Interleukin-1 Receptor-Associated Kinases , Interleukin-6/biosynthesis , JNK Mitogen-Activated Protein Kinases , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Ligands , Lipopolysaccharides/pharmacology , Lymphocytic choriomeningitis virus/physiology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Protein Kinases/genetics , Signal Transduction/drug effects , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/physiology , Toll-Like Receptors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...