Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 1605, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733557

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.


Assuntos
Anticorpos Neutralizantes/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Especificidade de Anticorpos , Humanos , Camundongos , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Conformação Proteica , Ratos
2.
Ann Rheum Dis ; 78(2): 228-237, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30459279

RESUMO

OBJECTIVE: Immune complexes (ICs) play a critical role in the pathology of autoimmune diseases. The aim of this study was to generate and characterise a first-in-class anti-FcγRIIA antibody (Ab) VIB9600 (previously known as MEDI9600) that blocks IgG immune complex-mediated cellular activation for clinical development. METHODS: VIB9600 was humanised and optimised from the IV.3 Ab. Binding affinity and specificity were determined by Biacore and ELISA. Confocal microscopy, Flow Cytometry-based assays and binding competition assays were used to assess the mode of action of the antibody. In vitro cell-based assays were used to demonstrate suppression of IC-mediated inflammatory responses. In vivo target suppression and efficacy was demonstrated in FcγRIIA-transgenic mice. Single-dose pharmacokinetic (PK)/pharmacodynamic study multiple dose Good Laboratory Practice (GLP) toxicity studies were conducted in non-human primates. RESULTS: We generated a humanised effector-deficient anti-FcγRIIA antibody (VIB9600) that potently blocks autoantibody and IC-mediated proinflammatory responses. VIB9600 suppresses FcγRIIA activation by blocking ligand engagement and by internalising FcγRIIA from the cell surface. VIB9600 inhibits IC-induced type I interferons from plasmacytoid dendritic cells (involved in SLE), antineutrophil cytoplasmic antibody (ANCA)-induced production of reactive oxygen species by neutrophils (involved in ANCA-associated vasculitis) and IC-induced tumour necrosis factor α and interleukin-6 production (involved in rheumatoid arthritis). In FcγRIIA transgenic mice, VIB9600 suppressed antiplatelet antibody-induced thrombocytopaenia, acute anti-GBM Ab-induced nephritis and anticollagen Ab-induced arthritis. VIB9600 also exhibited favourable PK and safety profiles in cynomolgus monkey studies. CONCLUSIONS: VIB9600 is a specific humanised antibody antagonist of FcγRIIA with null effector function that warrants further clinical development for the treatment of IC-mediated diseases.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Complexo Antígeno-Anticorpo/efeitos dos fármacos , Doenças Autoimunes/tratamento farmacológico , Fatores Imunológicos/farmacologia , Receptores de IgG/imunologia , Animais , Anticorpos Anticitoplasma de Neutrófilos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Doenças Autoimunes/imunologia , Células Dendríticas/imunologia , Humanos , Imunoglobulina G/imunologia , Interleucina-6/imunologia , Macaca fascicularis , Camundongos , Camundongos Transgênicos , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/imunologia , Fator de Necrose Tumoral alfa/imunologia
3.
Nat Commun ; 9(1): 1758, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717110

RESUMO

Although the aetiology of systemic lupus erythematosus (SLE) is unclear, dysregulated B cell responses have been implicated. Here we show that an unusual CD11chiT-bet+ B cell subset, with a unique expression profile including chemokine receptors consistent with migration to target tissues, is expanded in SLE patients, present in nephrotic kidney, enriched for autoreactive specificities and correlates with defined clinical manifestations. IL-21 can potently induce CD11chiT-bet+ B cells and promote the differentiation of these cells into Ig-secreting autoreactive plasma cells. While murine studies have identified a role for T-bet-expressing B cells in autoimmunity, this study describes and exemplifies the importance of CD11chiT-bet+ B cells in human SLE.


Assuntos
Linfócitos B/imunologia , Antígeno CD11c/imunologia , Diferenciação Celular/fisiologia , Interleucinas/fisiologia , Lúpus Eritematoso Sistêmico/metabolismo , Plasmócitos/citologia , Proteínas com Domínio T/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Subpopulações de Linfócitos B , Linfócitos B/metabolismo , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Plasmócitos/imunologia , Adulto Jovem
4.
Sci Transl Med ; 8(361): 361ra137, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798262

RESUMO

Autoantibodies can be present years to decades before the onset of disease manifestations in autoimmunity. This finding suggests that the initial autoimmune trigger involves a peripheral lymphoid component, which ultimately drives disease pathology in local tissues later in life. We show that Sjögren's syndrome manifestations that develop in aged NOD.H-2h4 mice were driven by and dependent on peripheral dysregulation that arose in early life. Specifically, elimination of spontaneous germinal centers in spleens of young NOD.H-2h4 mice by transient blockade of CD40 ligand (CD40L) or splenectomy abolished Sjögren's pathology of aged mice. Strikingly, a single injection of anti-CD40L at 4 weeks of age prevented tertiary follicle neogenesis and greatly blunted the formation of key autoantibodies implicated in glandular pathology, including anti-muscarinic receptor antibodies. Microarray profiling of the salivary gland characterized the expression pattern of genes that increased with disease progression and showed that early anti-CD40L greatly repressed B cell function while having a broader effect on multiple biological pathways, including interleukin-12 and interferon signaling. A single prophylactic treatment with anti-CD40L also inhibited the development of autoimmune thyroiditis and diabetes in NOD.H-2h4 and nonobese diabetic mice, respectively, supporting a key role for CD40L in the pathophysiology of several autoimmune models. These results strongly suggest that early peripheral immune dysregulation gives rise to autoimmune manifestations later in life, and for diseases predated by autoantibodies, early prophylactic intervention with biologics may prove efficacious.


Assuntos
Autoimunidade , Antígenos CD40/metabolismo , Sistema Imunitário/patologia , Envelhecimento , Animais , Anticorpos Monoclonais/farmacologia , Autoanticorpos/imunologia , Células da Medula Óssea/metabolismo , Antígenos CD40/genética , Ligante de CD40/antagonistas & inibidores , Ligante de CD40/imunologia , Citocinas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Modelos Animais de Doenças , Feminino , Ligantes , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Glândulas Salivares/metabolismo , Síndrome de Sjogren/genética , Síndrome de Sjogren/imunologia , Baço/metabolismo , Tireoidite Autoimune/genética , Tireoidite Autoimune/imunologia
5.
Am J Pathol ; 185(11): 2949-68, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26348576

RESUMO

Remodeling of blood vessels and lymphatics are prominent features of sustained inflammation. Angiopoietin-2 (Ang2)/Tie2 receptor signaling and tumor necrosis factor-α (TNF)/TNF receptor signaling are known to contribute to these changes in airway inflammation after Mycoplasma pulmonis infection in mice. We determined whether Ang2 and TNF are both essential for the remodeling on blood vessels and lymphatics, and thereby influence the actions of one another. Their respective contributions to the initial stage of vascular remodeling and sprouting lymphangiogenesis were examined by comparing the effects of function-blocking antibodies to Ang2 or TNF, given individually or together during the first week after infection. As indices of efficacy, vascular enlargement, endothelial leakiness, venular marker expression, pericyte changes, and lymphatic vessel sprouting were assessed. Inhibition of Ang2 or TNF alone reduced the remodeling of blood vessels and lymphatics, but inhibition of both together completely prevented these changes. Genome-wide analysis of changes in gene expression revealed synergistic actions of the antibody combination over a broad range of genes and signaling pathways involved in inflammatory responses. These findings demonstrate that Ang2 and TNF are essential and synergistic drivers of remodeling of blood vessels and lymphatics during the initial stage of inflammation after infection. Inhibition of Ang2 and TNF together results in widespread suppression of the inflammatory response.


Assuntos
Infecções por Mycoplasma/patologia , Mycoplasma pulmonis/fisiologia , Ribonuclease Pancreático/antagonistas & inibidores , Transdução de Sinais , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Feminino , Inflamação , Linfangiogênese , Sistema Linfático/metabolismo , Sistema Linfático/patologia , Vasos Linfáticos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Mycoplasma/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Pericitos/patologia , Sistema Respiratório/metabolismo , Sistema Respiratório/patologia , Ribonuclease Pancreático/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
PLoS One ; 10(2): e0115828, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25706559

RESUMO

Release of endogenous damage associated molecular patterns (DAMPs), including members of the S100 family, are associated with infection, cellular stress, tissue damage and cancer. The extracellular functions of this family of calcium binding proteins, particularly S100A8, S100A9 and S100A12, are being delineated. They appear to mediate their functions via receptor for advanced glycation endproducts (RAGE) or TLR4, but there remains considerable uncertainty over the relative physiological roles of these DAMPs and their pattern recognition receptors. In this study, we surveyed the capacity of S100 proteins to induce proinflammatory cytokines and cell migration, and the contribution RAGE and TLR4 to mediate these responses in vitro. Using adenoviral delivery of murine S100A9, we also examined the potential for S100A9 homodimers to trigger lung inflammation in vivo. S100A8, S100A9 and S100A12, but not the S100A8/A9 heterodimer, induced modest levels of TLR4-mediated cytokine production from human PBMC. In contrast, for most S100s including S100A9, RAGE blockade inhibited S100-mediated cell migration of THP1 cells and major leukocyte populations, whereas TLR4-blockade had no effect. Intranasal administration of murine S100A9 adenovirus induced a specific, time-dependent predominately macrophage infiltration that coincided with elevated S100A9 levels and proinflammatory cytokines in the BAL fluid. Inflammatory cytokines were markedly ablated in the TLR4-defective mice, but unexpectedly the loss of TLR4 signaling or RAGE-deficiency did not appreciably impact the S100A9-mediated lung pathology or the inflammatory cell infiltrate in the alveolar space. These data demonstrate that physiological levels of S100A9 homodimers can trigger an inflammatory response in vivo, and despite the capacity of RAGE and TLR4 blockade to inhibit responses in vitro, the response is predominately independent of both these receptors.


Assuntos
Calgranulina B/farmacologia , Movimento Celular/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Knockout , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptor 4 Toll-Like/metabolismo
7.
Eur J Immunol ; 44(12): 3669-79, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25229885

RESUMO

Anaphylatoxin C5a released upon complement activation is associated with both acute and chronic inflammations such as gout. The pathogenesis of gout was identified as uric acid crystal deposition in the joints that activates inflammasome, leading to IL-1ß release. However, little is known about the interaction between complement activation and monosodium urate/uric acid (MSU) crystal-induced inflammasome activation or IL-1ß production. Here, we report that MSU crystal-induced proinflammatory cytokines/chemokines in human whole blood is predominantly regulated by C5a through its interaction with C5a receptor. C5a induces pro-IL-1ß and IL-1ß production in human primary monocytes, and potentiates MSU or cholesterol crystals in IL-1ß production. This potentiation is caspase-1 dependent and requires intracellular Ca(2+) mobilization, K(+) efflux, and cathepsin B activity. Our results provide insight into the role of C5a as an endogenous priming signal that is required for the initiation of uric acid crystal-induced IL-1ß production. C5a could potentially be a therapeutic target together with IL-1ß antagonists for the treatment of complement-dependent and inflammasome-associated diseases.


Assuntos
Antioxidantes/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Complemento C5a/imunologia , Interleucina-1beta/imunologia , Monócitos/imunologia , Ácido Úrico/farmacologia , Antioxidantes/efeitos adversos , Cálcio/imunologia , Sinalização do Cálcio/imunologia , Caspase 1/imunologia , Feminino , Humanos , Inflamassomos/imunologia , Masculino , Monócitos/patologia , Potássio/imunologia , Ácido Úrico/efeitos adversos
8.
Ann Rheum Dis ; 71(8): 1402-10, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22730375

RESUMO

BACKGROUND: Angiopoietin (Ang)-1 and Ang-2, and their shared receptor Tie2, are expressed in rheumatoid arthritis (RA) synovial tissue, but the cellular targets of Ang signalling and the relative contributions of Ang-1 and Ang-2 to arthritis are poorly understood. OBJECTIVES: To determine the cellular targets of Ang signalling in RA synovial tissue, and the effects of Ang-2 neutralisation in murine collagen-induced arthritis (CIA). METHODS: RA and psoriatic arthritis (PsA) synovial biopsies were examined for expression of Tie2 and activated phospho (p)-Tie2 by quantitative immunohistochemistry and immunofluorescent double staining. Human monocyte and macrophage Tie2 expression was determined by flow cytometry and quantitative PCR. Regulation of macrophage intracellular signalling pathways and gene expression were examined by immunoblotting and ELISA. CIA was assessed in mice treated with saline, control antibody, prednisolone or neutralising anti-Ang-2 antibody. RESULTS: Expression of synovial Tie2 and p-Tie2 was similar in RA and PsA. Tie2 activation in RA patient synovial tissue was predominantly localised in synovial macrophages and was expressed by human macrophage. Ang-1 and Ang-2 stimulated activation of multiple intracellular signalling pathways, and cooperated with tumour necrosis factor to induce macrophage interleukin 6 and macrophage inflammatory protein 1α production. Ang-2 selectively suppressed macrophage thrombospondin-2 production. Ang-2 neutralisation significantly decreased disease severity, synovial inflammation, neo-vascularisation and joint destruction in established CIA. CONCLUSIONS: The authors identify synovial macrophages as primary targets of Ang signalling in RA, and demonstrate that Ang-2 promotes the pro-inflammatory activation of human macrophages. Ang-2 makes requisite contributions to pathology in CIA, indicating that targeting Ang-2 may be of therapeutic benefit in the treatment of RA.


Assuntos
Angiopoietina-2/farmacologia , Artrite Experimental/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/genética , Angiopoietina-1/metabolismo , Angiopoietina-1/farmacologia , Angiopoietina-2/imunologia , Angiopoietina-2/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/imunologia , Artrite Psoriásica/metabolismo , Artrite Psoriásica/patologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Quimiocina CCL3/biossíntese , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/biossíntese , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Fosforilação , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Trombospondinas/biossíntese
9.
Mol Med ; 14(7-8): 374-82, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18475307

RESUMO

Multiple lines of evidence suggest that inhibition of Type I Interferons, including IFN-alpha, may provide a therapeutic benefit for autoimmune diseases. Using a chemical genomics approach integrated with cellular and in vivo assays, we screened a small compound library to identify modulators of IFN-alpha biological effects. A genomic fingerprint was developed from both ex vivo patient genomic information and in vitro gene modulation from IFN-alpha cell-based stimulation. A high throughput genomic-based screen then was applied to prioritize 268 small molecule inhibitors targeting 41 different intracellular signaling pathways. Active compounds were profiled further for their ability to inhibit the activation and differentiation of human monocytes using disease-related stimuli. Inhibitors targeting NF-kappaB or Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling emerged as "dissociated inhibitors" because they did not modulate IFN-alpha anti-viral effects against HSV-1 but potently inhibited other immune-related functions. This work describes a novel strategy to identify small molecule inhibitors for the treatment of autoimmune disorders.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Interferon-alfa/antagonistas & inibidores , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antivirais/antagonistas & inibidores , Células Cultivadas , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Feminino , Genoma Humano , Genômica/instrumentação , Genômica/métodos , Humanos , Fatores Imunológicos/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia
10.
Bioorg Med Chem Lett ; 16(16): 4339-44, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16759861

RESUMO

The synthesis, structure-activity relationship, in vivo activity, and metabolic profile for a series of triazolopyridine-oxazole based p38 inhibitors are described. The deficiencies of the lead structure in the series, CP-808844, were overcome by changes to the C4 aryl group and the triazole side-chain culminating in the identification of several potential clinical candidates.


Assuntos
Inibidores Enzimáticos/farmacologia , Oxazóis/química , Piridinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/química , Química Farmacêutica , Desenho de Fármacos , Inibidores Enzimáticos/química , Humanos , Concentração Inibidora 50 , Cinética , Modelos Químicos , Solubilidade , Relação Estrutura-Atividade , Triazóis/química
11.
Proc Natl Acad Sci U S A ; 100(15): 9044-9, 2003 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-12835414

RESUMO

Prostaglandin (PG)E2 is a potent mediator of pain and inflammation, and high levels of this lipid mediator are observed in numerous disease states. The inhibition of PGE2 production to control pain and to treat diseases such as rheumatoid arthritis to date has depended on nonsteroidal antiinflammatory agents such as aspirin. However, these agents inhibit the synthesis of all prostanoids. To produce biologically active PGE2, PGE synthases catalyze the isomerization of PGH2 into PGE2. Recently, several PGE synthases have been identified and cloned, but their role in inflammation is not clear. To study the physiological role of the individual PGE synthases, we have generated by targeted homologous recombination a mouse line deficient in microsomal PGE synthase 1 (mPGES1) on the inbred DBA/1lacJ background. mPGES1-deficient (mPGES1-/-) mice are viable and fertile and develop normally compared with wild-type controls. However, mPGES1-/- mice displayed a marked reduction in inflammatory responses compared with mPGES1+/+ mice in multiple assays. Here, we identify mPGES1 as the PGE synthase that contributes to the pathogenesis of collagen-induced arthritis, a disease model of human rheumatoid arthritis. We also show that mPGES1 is responsible for the production of PGE2 that mediates acute pain during an inflammatory response. These findings suggest that mPGES1 provides a target for the treatment of inflammatory diseases and pain associated with inflammatory states.


Assuntos
Inflamação/fisiopatologia , Oxirredutases Intramoleculares/deficiência , Dor/fisiopatologia , Animais , Artrite Experimental/etiologia , Artrite Experimental/patologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/etiologia , Artrite Reumatoide/patologia , Artrite Reumatoide/fisiopatologia , Dinoprostona/biossíntese , Feminino , Humanos , Hipersensibilidade Tardia , Mediadores da Inflamação/metabolismo , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/fisiologia , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Dor/tratamento farmacológico , Prostaglandina-E Sintases
12.
Infect Immun ; 70(11): 6147-57, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379692

RESUMO

Organisms within the Hardjo serovar of Leptospira species are harbored in cattle throughout the world, causing abortion in pregnant animals as well as being shed in the urine, thereby providing sources of zoonotic infection for humans. We recently showed that sterile immunity in vaccinated cattle is associated with induction of a type 1 (Th1) cell-mediated immune response. Here naïve and previously vaccinated pregnant cattle were challenged with a virulent strain of serovar Hardjo and subsequently evaluated for expression of a type 1 immune response. Lymphocytes that responded in a recall response to antigen by undergoing blast transformation were evident in cultures of peripheral blood mononuclear cells (PBMC) from vaccinated cattle throughout the postchallenge test period while those from naïve cattle were evident at one time point only. Nevertheless, beginning at 2 weeks after challenge, gamma interferon (IFN-gamma) was measured in supernatants of antigen-stimulated PBMC cultures from nonvaccinated animals although the amount produced was always less than that in cultures of PBMC from vaccinated animals. IFN-gamma(+) cells were also evident in antigen-stimulated cultures of PBMC from vaccinated but not from nonvaccinated animals throughout the postchallenge period. The IFN-gamma(+) cells included CD4(+) and WC1(+) gammadelta T cells, and a similar proportion of these two subpopulations were found among the dividing cells in antigen-stimulated cultures as ascertained by carboxyfluorescein succinimidyl ester loading. Finally, while naïve and vaccinated animals had similar levels of antigen-specific immunoglobulin G1 (IgG1) following challenge, vaccinated animals had twofold-more IgG2. In conclusion, while infection may induce a type 1 response we suggest that it is too weak to prevent establishment of chronic infection.


Assuntos
Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Leptospira/imunologia , Glicoproteínas de Membrana/análise , Receptores de Antígenos de Linfócitos T gama-delta/análise , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Bovinos , Citometria de Fluxo , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Interferon gama/biossíntese , Vacinação
13.
Immunology ; 105(2): 181-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11872093

RESUMO

Bovine gammadelta T cells are stimulated to proliferate by autologous monocytes. This is referred to as the autologous mixed leucocyte reaction (AMLR). It has been shown previously that the stimulatory component is constitutively expressed on the monocyte plasma membrane and is a protein or has a protein moiety. Here we showed that gammadelta T-cell responses to the monocytes requires interaction with the T-cell receptor because Fab1 fragments of a monoclonal antibody (mAb) that reacts with the delta chain of the T-cell receptor blocked proliferation in the AMLR. Monocyte molecules involved in stimulation were also characterized further by biochemical and immunological methods. A mAb, named M5, was generated by immunizing mice with bovine monocytes and shown to block the ability of monocytes to stimulate in the AMLR. Treatment of monocytes or monocyte membranes with high salt, chelating agents or phospholipase C did not affect their ability to stimulate gammadelta T-cell proliferation or reactivity with mAb M5 indicating the ability of monocytes to stimulate does not involve peripheral membrane components or a glycosyl-phosphatidylinsositol (GPI)-anchored components. Hence it was concluded that the stimulation occurred as a result of intergral membrane proteins including that recognized by mAb M5. The ligand for mAb M5 was on all bovine monocytes and to a lower level on granulocytes but not on lymphocytes. MAb M5 also reacted with sheep monocytes but not with human monocytes or murine macrophages, in agreement with a previous reports that sheep monocytes but not human or mouse mononuclear phagocytes have the capacity to stimulate bovine gammadelta T cells in in vitro cultures. The level of expression of the M5 ligand was not altered by gamma-irradiation or culture of monocytes with lipopolysaccharide but it was decreased following culture with interferon-gamma-containing cell culture supernatants.


Assuntos
Monócitos/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/análise , Subpopulações de Linfócitos T/imunologia , Animais , Anticorpos Monoclonais/imunologia , Bovinos , Divisão Celular/imunologia , Células Cultivadas , Feminino , Humanos , Interferon gama/imunologia , Ligantes , Ativação Linfocitária/imunologia , Teste de Cultura Mista de Linfócitos , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Antígenos de Linfócitos T gama-delta/antagonistas & inibidores , Ovinos , Especificidade da Espécie
14.
Vet Immunol Immunopathol ; 84(3-4): 127-42, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11777529

RESUMO

Four fragments of the bovine IL-12 receptor beta2 were sequenced following generation by reverse transcriptase polymerase chain reaction (RT-PCR) amplification of RNA from mitogen-activated bovine peripheral blood mononuclear cells (PBMC). Primers were based on sequences within regions of the human IL-12Rbeta2 gene that displayed high levels of similarity with the mouse IL-12Rbeta2 gene sequence. The amplified bovine IL-12Rbeta2 fragments had 82-87% similarity at the nucleotide level with human IL-12Rbeta2 and 70-88% similarity at the predicted amino acid level. Bovine IL-12Rbeta2 gene expression was induced following culture of PBMC with Concanavalin A (Con A), with immobilized monoclonal antibody to CD3 or with human recombinant IL-12 p70 and correlated with interferon-gamma (IFN-gamma) production. Expression of bovine IL-12Rbeta2 by PBMC was detected by 2h of culture with Con A and sustained for at least 5 days when cultured with rHuIL-12. Expression, however, did not require cellular proliferation since IL-12 did not induce proliferation, although both Con A and anti-CD3 monoclonal antibody did do so. Addition of rHuIL-10 inhibited IFN-gamma production without abrogating bovine IL-12Rbeta2 gene expression.


Assuntos
Receptores de Interleucina/genética , Animais , Anticorpos Monoclonais/farmacologia , Sequência de Bases , Complexo CD3 , Bovinos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Concanavalina A/farmacologia , Primers do DNA/genética , DNA Complementar/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/biossíntese , Interleucina-10/farmacologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Receptores de Interleucina-12 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...