Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Arthritis Rheum ; 64(5): 1601-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22127978

RESUMO

OBJECTIVE: To examine the role of interferon regulatory factor 3 (IRF-3) in the regulation of interleukin-23 (IL-23) production in patients with systemic lupus erythematosus (SLE). METHODS: Bone marrow-derived macrophages were isolated from both wild-type and IRF3(-/-) C57BL/6 mice. These cells were stimulated with the Toll-like receptor 3 (TLR-3) agonist poly(I-C), and IL-23p19 cytokine levels were analyzed by enzyme-linked immunosorbent assay. IRF-3 binding to the IL-23p19 gene promoter region in monocytes from patients with SLE and healthy control subjects was analyzed by chromatin immunoprecipitation (ChIP) assay. Luciferase reporter gene assays were performed to identify key drivers of IL-23p19 promoter activity. TANK-binding kinase 1 (TBK-1) protein levels were determined by Western blotting. RESULTS: ChIP assays demonstrated that IRF-3 was stably bound to the human IL-23p19 promoter in monocytes; this association increased following TLR-3 stimulation. Patients with SLE demonstrated increased levels of IRF-3 bound to the IL-23p19 promoter compared with control subjects, which correlated with enhanced IL-23p19 production in monocytes from patients with SLE. Investigations of the TLR-3-driven responses in monocytes from patients with SLE revealed that TBK-1, which is critical for regulating IRF-3 activity, was hyperactivated in both resting and TLR-3-stimulated cells. CONCLUSION: Our results demonstrate for the first time that patients with SLE display enhanced IL-23p19 expression as a result of hyperactivation of TBK-1, resulting in increased binding of IRF-3 to the promoter. These findings provide novel insights into the molecular pathogenesis of SLE and the potential role for TLR-3 in driving this response.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Subunidade p19 da Interleucina-23/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/genética , Subunidade p19 da Interleucina-23/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Poli I-C/farmacologia , Análise Serial de Proteínas/métodos , Ligação Proteica , Proteínas Serina-Treonina Quinases/farmacologia , Receptor 3 Toll-Like/imunologia
2.
J Immunol ; 184(5): 2314-20, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20100929

RESUMO

Autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, result from a loss of tolerance to self-antigens and immune-mediated injury precipitated by the overproduction of type I IFN and inflammatory cytokines. We have identified the inositol 5' phosphatase SHIP-1 as a negative regulator of TLR3-induced type I IFN production. SHIP-1-deficient macrophages display enhanced TLR-induced IFN-beta production, and overexpression of SHIP-1 negatively regulates the ability of TLR3 and its adaptor, Toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta, to induce IFN-beta promoter activity, indicating that SHIP-1 negatively regulates TLR-induced IFN-beta production. Further dissection of the IFN-beta pathway implicates TANK-binding kinase 1 (TBK1) as the target for SHIP-1. Critically, in the absence of SHIP-1, TBK1 appears to be hyperphosphorylated both in unstimulated cells and following TLR3 stimulation. In addition, TBK1 appears to be constitutively associated with Toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta and TNFR-associated factor 3 in SHIP-1 deficient cells, whereas in wild-type cells this association is inducible following TLR3 stimulation. In support of a role for SHIP-1 in regulating complex formation, confocal microscopy demonstrates that TBK1 distribution in the cell is significantly altered in SHIP-1-deficient cells, with more prominent endosomal staining observed, compared with wild-type controls. Taken together, our results point to SHIP-1 as a critical negative regulator of IFN-beta production downstream of TLR3 through the regulation of TBK1 localization and activity.


Assuntos
Interferon beta/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor 3 Toll-Like/metabolismo , Aminoquinolinas/farmacologia , Animais , Western Blotting , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem Celular , Citocinas/metabolismo , Citometria de Fluxo , Humanos , Imiquimode , Mediadores da Inflamação/metabolismo , Inositol Polifosfato 5-Fosfatases , Interferon beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , NF-kappa B/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 3 Toll-Like/genética
3.
J Immunol ; 181(3): 1780-6, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18641315

RESUMO

Induction of type I IFNs is a fundamental cellular response to both viral and bacterial infection. The role of the transcription factor IRF3 is well established in driving this process. However, equally as important are cellular mechanisms for turning off type I IFN production to limit this response. In this respect, IRF3 has previously been shown to be targeted for ubiquitin-mediated degradation postviral detection to turn off the IFN-beta response. In this study, we provide evidence that the E3 ligase Ro52 (TRIM21) targets IRF3 for degradation post-pathogen recognition receptor activation. We demonstrate that Ro52 interacts with IRF3 via its C-terminal SPRY domain, resulting in the polyubiquitination and proteasomal degradation of the transcription factor. Ro52-mediated IRF3 degradation significantly inhibits IFN-beta promoter activity, an effect that is reversed in the presence of the proteasomal inhibitor MG132. Specific targeting of Ro52 using short hairpin RNA rescues IRF3 degradation following polyI:C-stimulation of HEK293T cells, with a subsequent increase in IFN-beta production. Additionally, shRNA targeting of murine Ro52 enhances the production of the IRF3-dependent chemokine RANTES following Sendai virus infection of murine fibroblasts. Collectively, this demonstrates a novel role for Ro52 in turning off and thus limiting IRF3-dependent type I IFN production by targeting the transcription factor for polyubiquitination and subsequent proteasomal degradation.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Interferon beta/biossíntese , Poliubiquitina/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Humanos , Fator Regulador 3 de Interferon/genética , Interferon beta/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores Imunológicos , Ribonucleoproteínas/genética , Ribonucleoproteínas/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
4.
PLoS One ; 9(7): e101503, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24999993

RESUMO

In recent years members of the tripartite motif-containing (TRIM) family of E3 ubiquitin ligases have been shown to both positively and negatively regulate viral defence and as such are emerging as compelling targets for modulating the anti-viral immune response. In this study we identify TRIM68, a close homologue of TRIM21, as a novel regulator of Toll-like receptor (TLR)- and RIG-I-like receptor (RLR)-driven type I IFN production. Proteomic analysis of TRIM68-containing complexes identified TRK-fused gene (TFG) as a potential TRIM68 target. Overexpression of TRIM68 and TFG confirmed their ability to associate, with TLR3 stimulation appearing to enhance the interaction. TFG is a known activator of NF-κB via its ability to interact with inhibitor of NF-κB kinase subunit gamma (IKK-γ) and TRAF family member-associated NF-κB activator (TANK). Our data identifies a novel role for TFG as a positive regulator of type I IFN production and suggests that TRIM68 targets TFG for lysosomal degradation, thus turning off TFG-mediated IFN-ß production. Knockdown of TRIM68 in primary human monocytes resulted in enhanced levels of type I IFN and TFG following poly(I:C) treatment. Thus TRIM68 targets TFG, a novel regulator of IFN production, and in doing so turns off and limits type I IFN production in response to anti-viral detection systems.


Assuntos
Autoantígenos/metabolismo , Imunidade Inata , Interferon beta/biossíntese , Proteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Vírus/imunologia , Autoantígenos/química , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Células HEK293 , Células HeLa , Humanos , Interferon beta/genética , Regiões Promotoras Genéticas/genética , Estrutura Terciária de Proteína , Proteólise , Receptores Imunológicos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/deficiência , Ubiquitinação
5.
J Exp Med ; 210(6): 1117-24, 2013 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-23690441

RESUMO

Retinoic acid (RA), a vitamin A metabolite, modulates mucosal T helper cell responses. Here we examined the role of RA in regulating IL-22 production by γδ T cells and innate lymphoid cells in intestinal inflammation. RA significantly enhanced IL-22 production by γδ T cells stimulated in vitro with IL-1ß or IL-18 and IL-23. In vivo RA attenuated colon inflammation induced by dextran sodium sulfate treatment or Citrobacter rodentium infection. This was associated with a significant increase in IL-22 secretion by γδ T cells and innate lymphoid cells. In addition, RA treatment enhanced production of the IL-22-responsive antimicrobial peptides Reg3ß and Reg3γ in the colon. The attenuating effects of RA on colitis were reversed by treatment with an anti-IL-22 neutralizing antibody, demonstrating that RA mediates protection by enhancing IL-22 production. To define the molecular events involved, we used chromatin immunoprecipitation assays and found that RA promoted binding of RA receptor to the IL-22 promoter in γδ T cells. Our findings provide novel insights into the molecular events controlling IL-22 transcription and suggest that one key outcome of RA signaling may be to shape early intestinal immune responses by promoting IL-22 synthesis by γδ T cells and innate lymphoid cells.


Assuntos
Colo/imunologia , Inflamação/imunologia , Interleucinas/imunologia , Linfócitos/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Tretinoína/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Citrobacter rodentium/imunologia , Colite/genética , Colite/imunologia , Colo/metabolismo , Sulfato de Dextrana/efeitos adversos , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/metabolismo , Inflamação/genética , Inflamação/metabolismo , Interleucinas/biossíntese , Interleucinas/genética , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores do Ácido Retinoico/imunologia , Receptores do Ácido Retinoico/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Transcrição Gênica/genética , Transcrição Gênica/imunologia , Tretinoína/metabolismo , Interleucina 22
7.
Cancer Res ; 72(3): 581-91, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22158905

RESUMO

The immunosuppressive microenvironment in tumors hampers the induction of antitumor immunity by vaccines or immunotherapies. Toll-like receptor (TLR) ligands have the potential to treat tumors, but they can exert a mixture of positive and negative effects on inflammation in the tumor microenvironment. In this study, we show that specific small molecule inhibitors of phosphoinositide 3-kinase (PI3K) relieve immunosuppression to heighten the proinflammatory effects of TLR ligands that support antitumor immunity. Multiple strategies to inhibit PI3K in dendritic cells (DC) each led to suppression of interleukin (IL)-10 and TGF-ß but did affect IL-12 or IL-1ß induction by the TLR5 ligand flagellin. In three different mouse models of cancer, combining flagellin with a class I PI3K inhibitor, either with or without a DC vaccine, delayed tumor growth and increased survival, with some animals exhibiting complete rejection and resistance to secondary challenge. Tumor growth suppression was associated with increased accumulation of polyfunctional T cells that secreted multiple effector cytokines, including IFN-γ, IL-17, and IL-2. Therapeutic protection was abolished in mice deficient in IL-17 or deprived of IFN-γ. Together, our results indicate that PI3K inhibition heighten the antitumor properties of TLR ligands, eliciting tumor regression directly but also indirectly by relieving suppressive signals that restrict potent antitumor T-cell responses. These findings suggest important uses for PI3K inhibitors in heightening responses to cancer immunotherapy and immunochemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia/métodos , Neoplasias Experimentais/terapia , Inibidores de Fosfoinositídeo-3 Quinase , Linfócitos T/metabolismo , Receptor 5 Toll-Like/agonistas , Animais , Western Blotting , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Neoplasias do Colo/terapia , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos/imunologia , Inibidores Enzimáticos/administração & dosagem , Flagelina/administração & dosagem , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tiazolidinedionas/administração & dosagem , Receptor 5 Toll-Like/metabolismo , Triazinas/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/imunologia
8.
Nat Rev Immunol ; 11(9): 617-25, 2011 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-21866173

RESUMO

The concept that viral sensing systems, via their ability to drive pro-inflammatory cytokine and interferon production, contribute to the development of autoimmune and autoinflammatory disease is supported by a wide range of clinical and experimental observations. Recently, the tripartite motif-containing proteins (TRIMs) have emerged as having key roles in antiviral immunity - either as viral restriction factors or as regulators of pathways downstream of viral RNA and DNA sensors, and the inflammasome. Given their involvement in these pathways, we propose that TRIM proteins contribute to the development and pathology of autoimmune and autoinflammatory conditions, thus making them potential novel targets for therapeutic manipulation.


Assuntos
Doenças Autoimunes/imunologia , Doenças Hereditárias Autoinflamatórias/imunologia , Inflamassomos/imunologia , Interferons/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Vírus/imunologia , Animais , Fatores de Restrição Antivirais , Doenças Autoimunes/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Interferons/metabolismo , Camundongos , Antígenos de Histocompatibilidade Menor , Domínios RING Finger , Proteínas Repressoras/metabolismo , Ribonucleoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Proteínas com Motivo Tripartido , Proteína 28 com Motivo Tripartido
9.
PLoS One ; 5(7): e11776, 2010 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-20668674

RESUMO

Ro52 is a member of the TRIM family of single-protein E3 ligases and is also a target for autoantibody production in systemic lupus erythematosus and Sjögren's syndrome. We previously demonstrated a novel function of Ro52 in the ubiquitination and proteasomal degradation of IRF3 following TLR3/4 stimulation. We now present evidence that Ro52 has a similar role in regulating the stability and activity of IRF7. Endogenous immunoprecipitation of Ro52-bound proteins revealed that IRF7 associates with Ro52, an effect which increases following TLR7 and TLR9 stimulation, suggesting that Ro52 interacts with IRF7 post-pathogen recognition. Furthermore, we show that Ro52 ubiquitinates IRF7 in a dose-dependent manner, resulting in a decrease in total IRF7 expression and a subsequent decrease in IFN-alpha production. IRF7 stability was increased in bone marrow-derived macrophages from Ro52-deficient mice stimulated with imiquimod or CpG-B, consistent with a role for Ro52 in the negative regulation of IRF7 signalling. Taken together, these results suggest that Ro52-mediated ubiquitination promotes the degradation of IRF7 following TLR7 and TLR9 stimulation. As Ro52 is known to be IFN-inducible, this system constitutes a negative-feedback loop that acts to protect the host from the prolonged activation of the immune response.


Assuntos
Fator Regulador 7 de Interferon/metabolismo , Ribonucleoproteínas/metabolismo , Receptores Toll-Like/metabolismo , Animais , Linhagem Celular , Células HeLa , Humanos , Imunoprecipitação , Fator Regulador 7 de Interferon/genética , Interferon-alfa/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , Ribonucleoproteínas/genética , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/genética , Ubiquitinação
10.
Dev Comp Immunol ; 33(4): 516-24, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19007808

RESUMO

The adaptive immune system is not completely developed when chickens hatch, so the innate immune system has evolved a range of mechanisms to deal with early pathogenic assault. Avian beta-defensins (AvBDs) and cathelicidins (CTHLs) are two major sub-classes of antimicrobial peptides (AMPs) with a fundamental role in both innate and adaptive immune responses. In this study, we demonstrate distinct expression patterns of innate immune genes including - Toll-like receptors (TLRs) (TLR2, TLR15 and TLR21, but not TLR4), the complete repertoire of AvBDs, CTHLs and both pro- and anti-inflammatory cytokines (IL1B, IL8, IFNG and IL10) during early chicken embryonic development. AvBD9 was significantly increased by over 150 fold at day 9; and AvBD10 was increased by over 100 fold at day 12 in the abdomen of the embryo, relative to day 3 expression levels (P<0.01). In contrast, AvBD14 was preferentially expressed in the head of the embryo. This is the first study to demonstrate differential patterns of AMP gene expression in the sterile environment of the developing embryo. Our results propose novel roles for AMPs during development and reveal the innate preparedness of developing embryos for pathogenic assault in ovo, or post-hatching.


Assuntos
Catelicidinas/genética , Embrião de Galinha/imunologia , Desenvolvimento Embrionário/imunologia , Expressão Gênica/imunologia , beta-Defensinas/genética , Animais , Catelicidinas/classificação , Embrião de Galinha/microbiologia , Citocinas/genética , Citocinas/imunologia , Desenvolvimento Embrionário/genética , Imunidade Inata/genética , Filogenia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , beta-Defensinas/classificação
11.
J Exp Med ; 206(8): 1661-71, 2009 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-19635858

RESUMO

Ro52/Trim21 is targeted as an autoantigen in systemic lupus erythematosus and Sjögren's syndrome. Polymorphisms in the Ro52 gene have been linked to these autoimmune conditions, but the molecular mechanism by which Ro52 may promote development of systemic autoimmune diseases has not been explored. To address this issue, we generated Ro52-null mice (Ro52(-/-)), which appear phenotypically normal if left unmanipulated. However, Ro52(-/-) mice develop severe dermatitis extending from the site of tissue injury induced by ear tags. The affected mice further develop several signs of systemic lupus with hypergammaglobulinemia, autoantibodies to DNA, proteinuria, and kidney pathology. Ro52, which was recently identified as an E3 ligase, mediates ubiquitination of several members of the interferon regulatory factor (IRF) family, and the Ro52-deficient mice have an enhanced production of proinflammatory cytokines that are regulated by the IRF transcription factors, including cytokines involved in the Th17 pathway (interleukin [IL] 6, IL-12/IL-23p40, and IL-17). Loss of IL-23/IL-17 by genetic deletion of IL-23/p19 in the Ro52(-/-) mice conferred protection from skin disease and systemic autoimmunity. These data reveal that the lupus-associated Ro52 protein is an important negative regulator of proinflammatory cytokine production, and they provide a mechanism by which a defective Ro52 function can lead to tissue inflammation and systemic autoimmunity through the IL-23-Th17 pathway.


Assuntos
Doenças Autoimunes/etiologia , Inflamação/etiologia , Interleucina-23/metabolismo , Ribonucleoproteínas/deficiência , Animais , Doenças Autoimunes/patologia , Sequência de Bases , Citocinas/biossíntese , Primers do DNA/genética , Modelos Animais de Doenças , Retroalimentação Fisiológica , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Interferons/farmacologia , Interleucina-23/deficiência , Interleucina-23/genética , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ribonucleoproteínas/genética , Subpopulações de Linfócitos T/classificação , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/classificação , Linfócitos T Auxiliares-Indutores/imunologia
12.
Biochem Soc Trans ; 36(Pt 3): 453-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18481980

RESUMO

The IRF [IFN (interferon) regulatory factor] family of transcription factors control many cellular processes, including induction of key antiviral cytokines, type I IFNs, following viral infection. Recent studies have revealed several endogenous and viral proteins involved in ubiquitin-mediated regulation of IRF activity and thus having an impact on type I IFN signalling. Through the ubiquitin pathway, these proteins can manipulate the antiviral response either by initiating proteasomal degradation of the IRFs or, in contrast, by promoting activation of the IRFs.


Assuntos
Antivirais/metabolismo , Fatores Reguladores de Interferon/metabolismo , Ubiquitinação , Sequência de Aminoácidos , Animais , Humanos , Fatores Reguladores de Interferon/química , Dados de Sequência Molecular , Ubiquitina/metabolismo
13.
Immunogenetics ; 59(7): 573-80, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17483936

RESUMO

Antimicrobial peptides (AMPs), essential components of innate immunity, are found in a range of phylogenetically diverse species and are thought to act by disrupting the membrane integrity of microbes. In this paper, we used evolutionary signatures to identify sites that are most relevant during the functional evolution of these molecules and introduced amino acid substitutions to improve activity. We first demonstrate that the anti-microbial activity of chicken avian beta-defensin-8, previously known as gallinacin-12, can be significantly increased against Escherichia coli, Listeria monocytogenes, Salmonella typhimurium, Salmonella typhimurium phoP- mutant and Streptococcus pyogenes through targeted amino acid substitutions, which confer increased peptide charge. However, by increasing the AMP charge through amino acid substitutions at sites predicted to be subject to positive selection, antimicrobial activity against Escherichia coli was further increased. In contrast, no further increase in activity was observed against the remaining pathogens. This result suggests that charge-increasing modifications confer increased broad-spectrum activity to an AMP, whilst positive selection at particular sites is involved in directing the antimicrobial response against specific pathogens. Thus, there is potential for the rational design of novel therapeutics based on specifically targeted and modified AMPs.


Assuntos
Substituição de Aminoácidos/genética , Proteínas Aviárias/genética , Evolução Molecular , beta-Defensinas/genética , Sequência de Aminoácidos , Substituição de Aminoácidos/imunologia , Animais , Proteínas Aviárias/fisiologia , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Galinhas , Relação Dose-Resposta Imunológica , Humanos , Dados de Sequência Molecular , beta-Defensinas/fisiologia
14.
Nat Rev Cardiol ; 8(1): 2, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21218552
15.
Nat Rev Cardiol ; 8(2): 62, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21348138
16.
19.
Infect Immun ; 74(3): 1692-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16495540

RESUMO

Toll-like receptors (TLRs) are a group of highly conserved molecules that initiate the innate immune response to pathogens by recognizing structural motifs expressed by microbes. We have identified a novel TLR, TLR15, by bioinformatic analysis of the chicken genome, which is distinct from any known vertebrate TLR and thus appears to be avian specific. The gene for TLR15 was sequenced and is found on chromosome 3, and it has archetypal TIR and transmembrane domains and a distinctive arrangement of extracellular leucine-rich regions. mRNA for TLR15 was detected in the spleen, bursa, and bone marrow of healthy chickens, suggesting a role for this novel receptor in constitutive host defense. Following in vivo Salmonella enterica serovar Typhimurium infection, quantitative real-time PCR demonstrated significant upregulation of TLR15 in the cecum of infected chickens. Interestingly, similar induction of TLR2 expression following infection was also observed. In vitro studies revealed TLR15 upregulation in chicken embryonic fibroblasts stimulated with heat-killed S. enterica serovar Typhimurium. Collectively, these results suggest a role for the TLR in avian defense against bacterial infection. We hypothesize that TLR15 may represent an avian-specific TLR that has been either retained in chicken and lost in other taxa or gained in the chicken.


Assuntos
Doenças das Aves/metabolismo , Salmonelose Animal/metabolismo , Salmonella typhimurium/fisiologia , Receptores Toll-Like/biossíntese , Animais , Células Cultivadas , Filogenia , Salmonelose Animal/genética , Receptores Toll-Like/química , Receptores Toll-Like/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA