Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Immunol ; 185(2): 1222-9, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20543107

RESUMO

The development of atherosclerosis, a chronic inflammatory disease characterized by the formation of arterial fibrotic plaques, has been shown to be reduced by IL-33 in vivo. However, whether IL-33 can directly affect macrophage foam cell formation, a key feature of atherosclerotic plaques, has not been determined. In this study, we investigated whether IL-33 reduces macrophage foam cell accumulation in vivo and if IL-33 reduces their formation in vitro using THP-1 and primary human monocyte-derived macrophages. In Apolipoprotein E(-/-) mice fed on a high fat diet, IL-33 treatment significantly reduced the accumulation of macrophage-derived foam cells in atherosclerotic plaques. IL-33 also reduced macrophage foam cell formation in vitro by decreasing acetylated and oxidized low-density lipoprotein uptake, reducing intracellular total and esterified cholesterol content and enhancing cholesterol efflux. These changes were associated with IL-33-mediated reduction in the expression of genes involved in modified low-density lipoprotein uptake, such as CD36, and simultaneous increase in genes involved in cholesterol efflux, including Apolipoprotein E, thereby providing a mechanism for such an action for this cytokine. IL-33 also decreased the expression of key genes implicated in cholesterol esterification and triglyceride storage, including Acyl-CoA:cholesterol acyltransferase 1 and Adipocyte differentiation-related protein. Furthermore, using bone marrow-derived macrophages from ST2(-/-) mice, we demonstrate that the IL-33 receptor, ST2, is integral to the action of IL-33 on macrophage foam cell formation. In conclusion, IL-33 has a protective role in atherosclerosis by reducing macrophage foam cell formation suggesting that IL-33 maybe a potential therapeutic agent against atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Células Espumosas/efeitos dos fármacos , Interleucinas/farmacologia , Macrófagos/efeitos dos fármacos , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Colesterol/metabolismo , Ésteres do Colesterol/metabolismo , Feminino , Células Espumosas/metabolismo , Células Espumosas/patologia , Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacocinética , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
J Immunol ; 184(10): 5827-34, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20410491

RESUMO

TNF-like protein 1A (TL1A), a TNF superfamily cytokine that binds to death receptor 3 (DR3), is highly expressed in macrophage foam cell-rich regions of atherosclerotic plaques, although its role in foam cell formation has yet to be elucidated. We investigated whether TL1A can directly stimulate macrophage foam cell formation in both THP-1 and primary human monocyte-derived macrophages with the underlying mechanisms involved. We demonstrated that TL1A promotes foam cell formation in human macrophages in vitro by increasing both acetylated and oxidized low-density lipoprotein uptake, by enhancing intracellular total and esterified cholesterol levels and reducing cholesterol efflux. This imbalance in cholesterol homeostasis is orchestrated by TL1A-mediated changes in the mRNA and protein expression of several genes implicated in the uptake and efflux of cholesterol, such as scavenger receptor A and ATP-binding cassette transporter A1. Furthermore, through the use of virally delivered DR3 short-hairpin RNA and bone marrow-derived macrophages from DR3 knockout mice, we demonstrate that DR3 can regulate foam cell formation and contributes significantly to the action of TL1A in this process in vitro. We show, for the first time, a novel proatherogenic role for both TL1A and DR3 that implicates this pathway as a target for the therapeutic intervention of atherosclerosis.


Assuntos
Diferenciação Celular/imunologia , Células Espumosas/citologia , Células Espumosas/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Transdução de Sinais/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Transporte Biológico/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Feminino , Células Espumosas/patologia , Humanos , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Regulação para Cima/imunologia
3.
J Neurosci ; 30(10): 3782-92, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20220013

RESUMO

Death receptor 3 is a proinflammatory member of the immunomodulatory tumor necrosis factor receptor superfamily, which has been implicated in several inflammatory diseases such as arthritis and inflammatory bowel disease. Intriguingly however, constitutive DR3 expression has been detected in the brains of mice, rats, and humans, although its neurological function remains unknown. By mapping the normal brain expression pattern of DR3, we found that DR3 is expressed specifically by cells of the neuron lineage in a developmentally regulated and region-specific pattern. Behavioral studies on DR3-deficient (DR3(ko)) mice showed that constitutive neuronal DR3 expression was required for stable motor control function in the aging adult. DR3(ko) mice progressively developed behavioral defects characterized by altered gait, dyskinesia, and hyperactivity, which were associated with elevated dopamine and lower serotonin levels in the striatum. Importantly, retrograde tracing showed that absence of DR3 expression led to the loss of corticostriatal innervation without significant neuronal loss in aged DR3(ko) mice. These studies indicate that DR3 plays a key nonredundant role in the retention of normal motor control function during aging in mice and implicate DR3 in progressive neurological disease.


Assuntos
Envelhecimento/fisiologia , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Destreza Motora/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/fisiologia , Envelhecimento/genética , Animais , Comunicação Celular/genética , Comunicação Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Corpo Estriado/crescimento & desenvolvimento , Corpo Estriado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurotransmissores/deficiência , Neurotransmissores/genética , Neurotransmissores/fisiologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/deficiência , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética
4.
Exp Eye Res ; 92(5): 338-43, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21296076

RESUMO

The interaction of the myeloid restricted molecule CD200R with its widely expressed ligand CD200 is involved in the down-regulation of microglia activation. In the present study, we examined the involvement of CD200R in microglia activation in experimental ocular hypertension to determine the role of microglia activation in retinal ganglion cell (RGC) death, the key pathological event in glaucoma. Experimental glaucoma was induced in adult Brown Norway rats by sclerosis of the episcleral veins with the injection of hypertonic saline. Immunohistochemical methods were used to determine the involvement of microglia using GFAP, CD45, OX42 and OX41 and the involvement of CD200 and CD200R in the optic nerve head. Our data demonstrate the increased presence of microglia within the optic nerve head during ocular hypertension, identified by positive staining with OX42 and OX41. The peak of microglia correlates with peak in RGC death at days 20-27 (T3) post OHT induction. In addition, CD200 and CD200R positive cells were increased in ocular hypertensive eyes. Increased expression of CD200 was detected in the early phase (days 1-7; T1) of OHT and decreased over time, whilst the expression of CD200R was detected in the middle phase (days 20-27; T3) of OHT, correlating with the increase in microglia markers. Changes in the expression of CD200R/CD200 occur early in experimental glaucoma and precede the peak in microglia infiltration and RGC death, suggesting that CD200R-positive microglia play an important role in the initiation of RGC death during OHT, indicating a potential area for therapeutic intervention in treating glaucoma.


Assuntos
Biomarcadores/metabolismo , Modelos Animais de Doenças , Glaucoma/metabolismo , Microglia/metabolismo , Receptores Imunológicos/metabolismo , Animais , Morte Celular , Técnica Indireta de Fluorescência para Anticorpo , Proteína Glial Fibrilar Ácida/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Masculino , Disco Óptico/metabolismo , Ratos , Ratos Endogâmicos BN , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
5.
Immunology ; 131(3): 340-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20518824

RESUMO

Macrophages (Mϕ) are professional antigen-presenting cells, but when they accumulate at sites of inflammation, they can inhibit T-cell proliferation. In experimental autoimmune uveoretinitis, this limits the expansion of T cells within the target organ. To define requirements for the elaboration of this outcome, we have generated populations of Mϕ in vitro that could also regulate T-cell responses; stimulating CD4(+) T-cell activation and cytokine production, but simultaneously suppressing T-cell proliferation. When T cells are removed from the influence of such cells, normal T-cell responses are restored. We show that tumour necrosis factor 1 (TNFR1) signalling is a critical checkpoint in the development of such Mϕ, as TNFR1(-/-) Mϕ are unable to suppress T-cell proliferation. This deficit in antigen-presenting cells results in a lack of production of prostaglandin E(2) (PGE(2)) and nitric oxide, which are critical effector mechanisms that inhibit T-cell division. However, TNFR1 signalling is not required for the inhibitory function of Mϕ because we could circumvent the requirement for this receptor, by maturing Mϕ in the presence of exogenous interferon-γ and PGE(2). This produced TNFR1(-/-) Mϕ that inhibited T-cell proliferation and indicates that TNFR1 delivers a signal that is necessary for the development but not the execution of this function.


Assuntos
Macrófagos/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Linfócitos T/metabolismo , Animais , Comunicação Celular , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Dinoprostona/genética , Dinoprostona/imunologia , Dinoprostona/metabolismo , Tolerância Imunológica , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
6.
Invest Ophthalmol Vis Sci ; 47(5): 2027-35, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16639012

RESUMO

PURPOSE: Experimental autoimmune uveoretinitis (EAU) can be induced in C57BL/6 mice (I-A(b)) using human retinoid-binding protein-3 (hRBP-3, previously IRBP) residues 1-20. This study of a truncated murine peptide (mRBP-3 1-16) was conducted to determine its pathogenic potential and to characterize partially its interaction with specific T cells. METHODS: After immunization with mRBP-3 1-16 or hRBP-3 1-20, EAU was assessed by immunohistochemistry. The immune response was assessed by tritiated thymidine incorporation and cytokine production analyzed by enzyme-linked immunosorbent assay (ELISA). T-cell receptor (TCR)- and major histocompatibility complex (MHC)-binding of mRBP-3 1-16 was studied by modeling and by using altered peptide ligands (APLs) and T-cell clones. RESULTS: mRBP-3 1-16 induced EAU in C57BL/6 mice, with severity and kinetics comparable to that after immunization with hRBP-3 1-20. T cells taken from mice immunized with mRBP-3 1-16 had a Th1 phenotype and proliferated in response to reactivation with mRBP-3 1-16, hRBP-3 1-20, or mRBP-3 1-16 APLs. mRBP-3 1-16 APLs elicited at least five distinct patterns of reactivity when tested with the mRBP-3 1-16-reactive T-cell clones. CONCLUSIONS: mRBP-3 1-16 immunizes and causes EAU in C57BL/6 mice. The studies using T-cell clones and APLs demonstrate that the immune response to mRBP-3 1-16 is drawn from a diverse population of antigen-specific T cells with a Th1 phenotype. Modeling and analysis of clones indicate that nonpathogenic T cells of an mRBP-3 1-16-reactive T-cell line recognize the peptide in a single register.


Assuntos
Doenças Autoimunes/etiologia , Proteínas do Olho/imunologia , Fragmentos de Peptídeos/imunologia , Retinite/etiologia , Proteínas de Ligação ao Retinol/imunologia , Uveíte/etiologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunização , Ligantes , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/imunologia , Retinite/imunologia , Retinite/patologia , Células Th1/imunologia , Uveíte/imunologia , Uveíte/patologia
7.
J Immunol Methods ; 287(1-2): 67-77, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15099756

RESUMO

Respiratory tract dendritic cells (RTDC) form a contiguous subepithelial network within the nasorespiratory tract bridging innate and acquired immunity and have been implicated in nasal mucosal tolerance induction. Discrepancies exist between isolation techniques with respect to phenotype and function of RTDC. Therefore, the aim of this study was to modify previous methods to provide a consistent isolation method whilst maintaining good cell viability and enriched cell numbers so as to facilitate further phenotype and functional studies of murine RTDC. RTDCs isolated by enzyme digestion, Percoll density gradient centrifugation and overnight GM-CSF culture followed by MACS separation retain an archetypical immature dendritic cell phenotype, characterised by MHCII(low) CD40(neg) CD86(neg) CD80(neg) CD11c(low) cell surface expression. Splenic-derived DC (SDC) isolated conformed to a day 1 in vitro phenotype; MHCII(low) CD40(neg) CD86(low) CD80(neg) CD11c(low) but can further mature phenotypically in vitro. Both RTDC and SDC processed and presented antigen efficiently to T cells in vitro. Using such modified isolation procedures for RTDCs, we have developed a consistent method of RTDC enrichment, which maintains the immature phenotype and functional antigen presenting capability.


Assuntos
Separação Celular/métodos , Células Dendríticas/imunologia , Sistema Respiratório/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Cultivadas , Técnicas de Cocultura , Citocinas/análise , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Fenótipo , Sistema Respiratório/citologia , Sistema Respiratório/metabolismo , Baço/citologia , Baço/metabolismo , Linfócitos T/imunologia
9.
Expert Rev Clin Immunol ; 3(1): 73-83, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20476953

RESUMO

There has been longstanding interest in how B-cell responses may contribute to the pathology of neurological diseases. Traditionally, the premise has been that any such contribution relates to the potential of B cells to produce autopathogenic antibodies. Targeting autoantibodies continues to be an important therapeutic approach, particularly in disorders where the role of antibodies is relatively well established, such as in certain inflammatory disorders of peripheral nerves or the neuromuscular junction. In other conditions, such as multiple sclerosis, the role of circulating antibodies targeting the CNS has been less clear, although pathologic studies continue to implicate CNS-reactive antibodies, as well as B cells within the CNS compartment. Recently, new insights into fundamental properties of B cells have suggested that these cells may contribute in an antibody-independent fashion, both to normal immune responses, and in the context of immune mediated diseases. Here, we will consider the potential roles of antibody-dependent as well as antibody-independent B-cell involvement in multiple sclerosis. The topic is of particular interest at a time that B-cell-directed therapies are being evaluated for this and other autoimmune diseases.

10.
Am J Pathol ; 171(2): 580-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17600119

RESUMO

Macrophage responses are regulated by multiple secreted factors as well as by cell surface receptors, including the inhibitory signals resulting from ligation of myeloid CD200 receptors (CD200R) by the widely distributed CD200. In the absence of CD200, animals display increased susceptibility to autoimmunity and earlier onset aggressive autoimmune disease. In these current experiments, an agonist monoclonal rat anti-mouse CD200R (DX109) antibody delivered a negative signal to bone marrow-derived macrophages, which suppressed interferon (IFN)gamma-mediated nitric oxide (NO) and interleukin-6 production. Experimental autoimmune uveoretinitis (EAU) was used as a model of organ-specific autoimmunity in the eye, a tissue with extensive neuronal and endothelial CD200 expression. In mice lacking CD200 (CD200(-/-)), increased numbers of retina-infiltrating macrophages displaying heightened NO responses were observed during EAU. In addition, we aimed to suppress disease by maintaining tonic suppression of macrophage activation via CD200R. Systemically administered DX109 monoclonal antibody suppressed EAU despite maintained T-cell proliferation and IFNgamma production. Furthermore, locally administered DX109 monoclonal antibody resulted in an earlier resolution of disease. These experiments demonstrate that promoting CD200R-mediated signaling can successfully prevent full expression of IFNgamma-mediated macrophage activation and protect against tissue damage during autoimmune responses.


Assuntos
Anticorpos Monoclonais/farmacologia , Doenças Autoimunes/prevenção & controle , Ativação de Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/imunologia , Retinite/prevenção & controle , Uveíte/prevenção & controle , Animais , Anticorpos Monoclonais/imunologia , Doenças Autoimunes/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Relação Dose-Resposta a Droga , Interferon gama/metabolismo , Interferon gama/farmacologia , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Células Mieloides/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Ratos , Retinite/patologia , Transdução de Sinais/imunologia , Fatores de Tempo , Receptores Toll-Like/metabolismo , Uveíte/patologia
11.
Methods ; 38(2): 69-76, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16414268

RESUMO

Delivering soluble (auto) antigenic peptides via the naso-respiratory route induces tolerance to that peptide and suppression of experimental models of autoimmune disease. In the normal lung, respiratory tract dendritic cells (RTDCs) efficiently endocytose soluble antigens, migrate to regional lymph nodes and present peptide to T cells that subsequently become tolerant. This article describes protocols for inducing tolerance via the naso-respiratory tract in experimental autoimmune uveoretinitis (EAU); for the isolation of RTDCs to facilitate definition of, and conditions for, maturation and activation of cells; and to test RTDC ability to induce tolerance in murine EAU when adoptively transferred.


Assuntos
Doenças Autoimunes/terapia , Oftalmopatias/terapia , Tolerância Imunológica/imunologia , Imunoterapia/métodos , Mucosa Nasal/imunologia , Transferência Adotiva/métodos , Animais , Antígenos de Superfície/imunologia , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/patologia , Linfócitos T CD4-Positivos/imunologia , Separação Celular , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/transplante , Oftalmopatias/induzido quimicamente , Oftalmopatias/patologia , Proteínas do Olho/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Imunidade nas Mucosas/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Sistema Respiratório/citologia , Sistema Respiratório/imunologia , Retinite/induzido quimicamente , Retinite/patologia , Retinite/terapia , Proteínas de Ligação ao Retinol/imunologia , Baço/citologia , Baço/imunologia , Baço/metabolismo , Uveíte/induzido quimicamente , Uveíte/patologia , Uveíte/terapia
12.
J Immunol ; 175(10): 6286-93, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16272279

RESUMO

IFN-gamma stimulates macrophage activation and NO production, which leads to destruction of the retina in experimental autoimmune uveoretinitis. In this study, we investigate the mechanism of disease resistance in TNF p55 receptor-deficient animals. We show that although T cell priming is relatively unaffected, macrophages lacking the TNF p55 receptor fail to produce NO following IFN-gamma stimulation because of a requirement for autocrine TNF-alpha signaling through the TNF p55 receptor. In contrast to the impaired activation of NO synthesis, MHC class II up-regulation was indistinguishable in wild-type and TNFRp55-/- mice stimulated with IFN-gamma. These defects could be overcome by stimulating macrophages with LPS. Together, these results show that selected aspects of IFN-gamma activation are controlled by autocrine secretion of TNF-alpha, but that this control is lost in the presence of signals generated by pathogen-associated molecular patterns recognizing receptors.


Assuntos
Doenças Autoimunes/imunologia , Interferon gama/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Retinite/imunologia , Uveíte/imunologia , Animais , Comunicação Autócrina , Antígenos CD40/metabolismo , Feminino , Antígenos de Histocompatibilidade Classe II/metabolismo , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA