Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 26(11): 2753-64, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25788530

RESUMO

The myeloid differentiation protein 88 (MyD88) adapter protein is an important mediator of kidney allograft rejection, yet the precise role of MyD88 signaling in directing the host immune response toward the development of kidney allograft rejection remains unclear. Using a stringent mouse model of allogeneic kidney transplantation, we demonstrated that acute allograft rejection occurred equally in MyD88-sufficient (wild-type [WT]) and MyD88(-/-) recipients. However, MyD88 deficiency resulted in spontaneous diminution of graft infiltrating effector cells, including CD11b(-)Gr-1(+) cells and activated CD8 T cells, as well as subsequent restoration of near-normal renal graft function, leading to long-term kidney allograft acceptance. Compared with T cells from WT recipients, T cells from MyD88(-/-) recipients failed to mount a robust recall response upon donor antigen restimulation in mixed lymphocyte cultures ex vivo. Notably, exogenous IL-6 restored the proliferation rate of T cells, particularly CD8 T cells, from MyD88(-/-) recipients to the proliferation rate of cells from WT recipients. Furthermore, MyD88(-/-) T cells exhibited diminished expression of chemokine receptors, specifically CCR4 and CXCR3, and the impaired ability to accumulate in the kidney allografts despite an otherwise MyD88-sufficient environment. These results provide a mechanism linking the lack of intrinsic MyD88 signaling in T cells to the effective control of the rejection response that results in spontaneous resolution of acute rejection and long-term graft protection.


Assuntos
Rejeição de Enxerto , Síndromes de Imunodeficiência/genética , Transplante de Rim , Rim/imunologia , Fator 88 de Diferenciação Mieloide/genética , Aloenxertos , Animais , Antígeno CD11b/metabolismo , Proliferação de Células , Sobrevivência de Enxerto , Interleucina-6/metabolismo , Rim/patologia , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Knockout , Doenças da Imunodeficiência Primária , Receptores CCR4/metabolismo , Receptores CXCR3/metabolismo , Transdução de Sinais , Transplante de Pele , Linfócitos T/citologia , Transplante Homólogo
2.
Pigment Cell Melanoma Res ; 27(6): 1075-85, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24935676

RESUMO

To generate a mouse model of spontaneous epidermal depigmentation, parental h3TA2 mice, expressing both a human-derived, tyrosinase-reactive T-cell receptor on T cells and the matching HLA-A2 transgene, were crossed to keratin 14-promoter driven, stem cell factor transgenic (K14-SCF) mice with intra-epidermal melanocytes. In resulting Vitesse mice, spontaneous skin depigmentation precedes symmetrical and sharply demarcated patches of graying hair. Whereas the SCF transgene alone dictates a greater retinoic acid receptor-related orphan receptor gamma (RORγt)(+) T-cell compartment, these cells displayed markedly increased IL-17 expression within Vitesse mice. Similar to patient skin, regulatory T cells were less abundant compared with K14-SCF mice, with the exception of gradually appearing patches of repigmenting skin. The subtle repigmentation observed likely reflects resilient melanocytes that coexist with skin-infiltrating, melanocyte-reactive T cells. Similar repigmenting lesions were found in a different TCR transgenic model of vitiligo developed on an SCF transgenic background, supporting a role for SCF in repigmentation.


Assuntos
Epiderme/patologia , Hipopigmentação/complicações , Hipopigmentação/imunologia , Imunidade , Pigmentação , Vitiligo/complicações , Vitiligo/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Epitopos , Humanos , Hipopigmentação/patologia , Interleucina-17/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monofenol Mono-Oxigenase/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Células-Tronco/genética , Linfócitos T Reguladores/imunologia , Transgenes , Vitiligo/patologia
3.
J Invest Dermatol ; 134(5): 1285-1294, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24366614

RESUMO

T-cell cytolytic activity targeting epidermal melanocytes is shown to cause progressive depigmentation and autoimmune vitiligo. By using the recently developed transgenic mice h3TA2 that carry T cells with a HLA-A2-restricted human tyrosinase peptide (h-Tyr)-reactive TCR and develop spontaneous vitiligo from an early age, we addressed the mechanism regulating autoimmune vitiligo. Depigmentation was significantly impaired only in IFN-γ-knockout h3TA2 mice but not in TNF-α- or perforin-knockout h3TA2 mouse strains, confirming a central role for IFN-γ in vitiligo development. In addition, regulatory T cells (Tregs) were relatively abundant in h3TA2-IFN-γ(-/-) mice, and depletion of the Treg-engaging anti-CD25 antibody fully restored the depigmentation phenotype in h3TA2-IFN-γ(-/-) mice, mediated in part through the upregulation of proinflammatory cytokines such as IL-17 and IL-22. Further therapeutic potential of Treg abundance in preventing progressive depigmentation was evaluated by adoptively transferring purified Treg or using rapamycin. Both the adoptive transfer of Tregs and the use of rapamycin induced a lasting remission of vitiligo in mice treated at the onset of disease, or in mice with established disease. This leads us to conclude that reduced regulatory responses are pivotal to the development of vitiligo in disease-prone mice, and that a quantitative increase in the Treg population may be therapeutic for vitiligo patients with active disease.


Assuntos
Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Vitiligo/imunologia , Vitiligo/patologia , Transferência Adotiva , Animais , Autoimunidade/imunologia , Progressão da Doença , Células Epidérmicas , Epiderme/imunologia , Feminino , Humanos , Imunossupressores/farmacologia , Interferon gama/imunologia , Masculino , Melanócitos/citologia , Melanócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CCR5/genética , Receptores CXCR3/genética , Sirolimo/farmacologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/genética , Vitiligo/tratamento farmacológico
4.
J Clin Immunol ; 31(3): 379-94, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21287397

RESUMO

Tolerance therapy with nucleosomal histone peptides H4(71-94), H4(16-39), or H1'(22-42) controls disease in lupus-prone SNF1 mice. It would be clinically important to determine whether a cocktail of the above epitopes would be superior. Herein, we found that compared with cocktail peptides, H4(71-94) monotherapy more effectively delayed nephritis onset, prolonged lifespan, diminished immunoglobulin G autoantibody levels, reduced autoantigen-specific Th1 and Th17 responses and frequency of T(FH) cells in spleen and the helper ability of autoimmune T cells to B cells, by inducing potent CD8 Treg cells. H4(71-94) therapy was superior in "tolerance spreading," suppressing responses to other autoepitopes, nucleosomes, and ribonucleoprotein. We also developed an in vitro assay for therapeutic peptides (potentially in humans), which showed that H4(71-94), without exogenous transforming growth factor (TGF)-ß, was efficient in inducing stable CD4(+)CD25(+)Foxp3(+) T cells by decreasing interleukin 6 and increasing TGF-ß production by dendritic cells that induced ALK5-dependent Smad-3 phosphorylation (TGF-ß signal) in target autoimmune CD4(+) T cells.


Assuntos
Autoimunidade/efeitos dos fármacos , Histonas/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Imunoensaio , Fatores Imunológicos/farmacologia , Nefrite Lúpica/tratamento farmacológico , Peptídeos/farmacologia , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Combinação de Medicamentos , Epitopos/imunologia , Feminino , Citometria de Fluxo , Histonas/síntese química , Histonas/imunologia , Tolerância Imunológica/imunologia , Fatores Imunológicos/síntese química , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Transgênicos , Nucleossomos/imunologia , Nucleossomos/metabolismo , Peptídeos/síntese química , Peptídeos/imunologia
5.
Arthritis Res Ther ; 11(2): R59, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19405952

RESUMO

INTRODUCTION: Lupus patients need alternatives to steroids and cytotoxic drugs. We recently found that apigenin, a non-mutagenic dietary flavonoid, can sensitize recurrently activated, normal human T cells to apoptosis by inhibiting nuclear factor-kappa-B (NF-kappaB)-regulated Bcl-xL, cyclooxygenase 2 (COX-2), and cellular FLICE-like inhibitory protein (c-FLIP) expression. Because sustained immune activation and hyperexpression of COX-2 and c-FLIP contribute to lupus, we treated SNF1 mice that spontaneously develop human lupus-like disease with apigenin. METHODS: SNF1 mice with established lupus-like disease were injected with 20 mg/kg of apigenin daily and then monitored for development of severe nephritis. Histopathologic changes in kidneys, IgG autoantibodies to nuclear autoantigens in serum and in cultures of splenocytes, along with nucleosome-specific T helper 1 (Th1) and Th17 responses, COX-2 expression, and apoptosis of lupus immune cells were analyzed after apigenin treatment. RESULTS: Apigenin in culture suppressed responses of Th1 and Th17 cells to major lupus autoantigen (nucleosomes) up to 98% and 92%, respectively, and inhibited the ability of lupus B cells to produce IgG class-switched anti-nuclear autoantibodies helped by these Th cells in presence of nucleosomes by up to 82%. Apigenin therapy of SNF1 mice with established lupus suppressed serum levels of pathogenic autoantibodies to nuclear antigens up to 97% and markedly delayed development of severe glomerulonephritis. Apigenin downregulated COX-2 expression in lupus T cells, B cells, and antigen-presenting cells (APCs) and caused their apoptosis. Autoantigen presentation and Th17-inducing cytokine production by dendritic cells were more sensitive to the inhibitory effect of apigenin in culture, as evident at 0.3 to 3 muM, compared with concentrations (10 to 100 microM) required for inducing apoptosis. CONCLUSIONS: Apigenin inhibits autoantigen-presenting and stimulatory functions of APCs necessary for the activation and expansion of autoreactive Th1 and Th17 cells and B cells in lupus. Apigenin also causes apoptosis of hyperactive lupus APCs and T and B cells, probably by inhibiting expression of NF-kappaB-regulated anti-apoptotic molecules, especially COX-2 and c-FLIP, which are persistently hyperexpressed by lupus immune cells. Increasing the bioavailability of dietary plant-derived COX-2 and NF-kappaB inhibitors, such as apigenin, could be valuable for suppressing inflammation in lupus and other Th17-mediated diseases like rheumatoid arthritis, Crohn disease, and psoriasis and in prevention of inflammation-based tumors overexpressing COX-2 (colon, breast).


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Apigenina/farmacologia , Autoantígenos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Apoptose/efeitos dos fármacos , Autoanticorpos/biossíntese , Autoanticorpos/sangue , Autoanticorpos/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Ciclo-Oxigenase 2 , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Imunoglobulina G/efeitos dos fármacos , Interferon gama/biossíntese , Interferon gama/efeitos dos fármacos , Interleucina-17/imunologia , Nefrite Lúpica/imunologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos NZB , Nucleossomos/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia
6.
J Immunol ; 178(12): 7849-58, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548623

RESUMO

Subnanomolar doses of an unaltered, naturally occurring nucleosomal histone peptide epitope, H4(71-94), when injected s.c. into lupus-prone mice, markedly prolong lifespan by generating CD4+25+ and CD8+ regulatory T cells (Treg) producing TGF-beta. The induced Treg cells suppress nuclear autoantigen-specific Th and B cells and block renal inflammation. Splenic dendritic cells (DC) captured the s.c.-injected H4(71-94) peptide rapidly and expressed a tolerogenic phenotype. The DC of the tolerized animal, especially plasmacytoid DC, produced increased amounts of TGF-beta, but diminished IL-6 on stimulation via the TLR-9 pathway by nucleosome autoantigen and other ligands; and those plasmacytoid DC blocked lupus autoimmune disease by simultaneously inducing autoantigen-specific Treg and suppressing inflammatory Th17 cells that infiltrated the kidneys of untreated lupus mice. Low-dose tolerance with H4(71-94) was effective even though the lupus immune system is spontaneously preprimed to react to the autoepitope. Thus, H4(71-94) peptide tolerance therapy that preferentially targets pathogenic autoimmune cells could spare lupus patients from chronically receiving toxic agents or global immunosuppressants and maintain remission by restoring autoantigen-specific Treg cells.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Células Dendríticas/imunologia , Terapia de Imunossupressão , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/imunologia , Antígenos CD4/análise , Antígenos CD8/análise , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/metabolismo , Histonas/química , Histonas/imunologia , Tolerância Imunológica , Imunoglobulina G/imunologia , Interleucina-17/metabolismo , Subunidade alfa de Receptor de Interleucina-2/análise , Interleucina-6/metabolismo , Camundongos , Nucleossomos/imunologia , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Peptídeos/uso terapêutico , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
J Immunol ; 174(6): 3247-55, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15749855

RESUMO

We induced very low-dose tolerance by injecting lupus prone (SWR x NZB)F1 (SNF1) mice with 1 mug nucleosomal histone peptide autoepitopes s.c. every 2 wk. The subnanomolar peptide therapy diminished autoantibody levels and prolonged life span by delaying nephritis, especially by reducing inflammatory cell reaction and infiltration in kidneys. H4(71-94) was the most effective autoepitope. Low-dose tolerance therapy induced CD8+, as well as CD4+ CD25+ regulatory T (Treg) cell subsets containing autoantigen-specific cells. These adaptive Treg cells suppressed IFN-gamma responses of pathogenic lupus T cells to nucleosomal epitopes at up to a 1:100 ratio and reduced autoantibody production up to 90-100% by inhibiting nucleosome-stimulated T cell help to nuclear autoantigen-specific B cells. Both CD4+ CD25+ and CD8+ Treg cells produced and required TGF-beta1 for immunosuppression, and were effective in suppressing lupus autoimmunity upon adoptive transfer in vivo. The CD4+ CD25+ T cells were partially cell contact dependent, but CD8+ T cells were contact independent. Thus, low-dose tolerance with highly conserved histone autoepitopes repairs a regulatory defect in systemic lupus erythematosus by generating long-lasting, TGF-beta-producing Treg cells, without causing allergic/anaphylactic reactions or generalized immunosuppression.


Assuntos
Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/terapia , Proteínas Nucleares/imunologia , Nucleossomos/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Sequência de Aminoácidos , Animais , Anticorpos Antinucleares/sangue , Autoantígenos/administração & dosagem , Autoantígenos/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos/administração & dosagem , Epitopos/genética , Feminino , Tolerância Imunológica , Terapia de Imunossupressão , Lúpus Eritematoso Sistêmico/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Nefrite Lúpica/prevenção & controle , Nefrite Lúpica/terapia , Camundongos , Camundongos Endogâmicos NZB , Dados de Sequência Molecular , Proteínas Nucleares/genética , Peptídeos/imunologia , Receptores de Interleucina-2/metabolismo , Fator de Crescimento Transformador beta/biossíntese
8.
Nat Med ; 10(4): 411-5, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14991050

RESUMO

Autoimmune T-helper cells drive pathogenic autoantibody production in systemic lupus erythematosus (SLE), but the mechanisms maintaining those T cells are unknown. Autoreactive T cells are normally eliminated by functional inactivation (anergy) and activation-induced cell death (AICD) or apoptosis through death receptor (Fas) signaling. However, mutations in the genes encoding Fas and its ligand (FasL) are rare in classical SLE. By gene microarray profiling, validated by functional and biochemical studies, we establish here that activated T cells of lupus patients resist anergy and apoptosis by markedly upregulating and sustaining cyclooxygenase-2 (COX-2) expression. Inhibition of COX-2 caused apoptosis of the anergy-resistant lupus T cells by augmenting Fas signaling and markedly decreasing the survival molecule c-FLIP (cellular homolog of viral FLICE inhibitory protein). Studies with COX-2 inhibitors and Cox-2-deficient mice confirmed that this COX-2/FLIP antiapoptosis program is used selectively by anergy-resistant lupus T cells, and not by cancer cells or other autoimmune T cells. Notably, the gene encoding COX-2 is located in a lupus-susceptibility region on chromosome 1. We also found that only some COX-2 inhibitors were able to suppress the production of pathogenic autoantibodies to DNA by causing autoimmune T-cell apoptosis, an effect that was independent of prostaglandin E(2) (PGE(2)). These findings could be useful in the design of lupus therapies.


Assuntos
Isoenzimas/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Linfócitos T/imunologia , Regulação para Cima , Sequência de Bases , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/uso terapêutico , Primers do DNA , Feminino , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/enzimologia , Proteínas de Membrana , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Linfócitos T/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA