Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Science ; 376(6596): 996-1001, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35617401

RESUMO

T cell quiescence is essential for maintaining a broad repertoire against a large pool of diverse antigens from microbes and tumors, but the underlying molecular mechanisms remain largely unknown. We show here that CD8α is critical for the maintenance of CD8+ T cells in a physiologically quiescent state in peripheral lymphoid organs. Upon inducible deletion of CD8α, both naïve and memory CD8+ T cells spontaneously acquired activation phenotypes and subsequently died without exposure to specific antigens. PILRα was identified as a ligand for CD8α in both mice and humans, and disruption of this interaction was able to break CD8+ T cell quiescence. Thus, peripheral T cell pool size is actively maintained by the CD8α-PILRα interaction in the absence of antigen exposure.


Assuntos
Antígenos CD8 , Linfócitos T CD8-Positivos , Ativação Linfocitária , Glicoproteínas de Membrana , Receptores Imunológicos , Animais , Antígenos , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Deleção de Genes , Glicoproteínas de Membrana/metabolismo , Camundongos , Receptores Imunológicos/metabolismo
2.
Sci Transl Med ; 13(604)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321321

RESUMO

The immature and dysfunctional vascular network within solid tumors poses a substantial obstacle to immunotherapy because it creates a hypoxic tumor microenvironment that actively limits immune cell infiltration. The molecular basis underpinning this vascular dysfunction is not fully understood. Using genome-scale receptor array technology, we showed here that insulin-like growth factor binding protein 7 (IGFBP7) interacts with its receptor CD93, and we subsequently demonstrated that this interaction contributes to abnormal tumor vasculature. Both CD93 and IGFBP7 were up-regulated in tumor-associated endothelial cells. IGFBP7 interacted with CD93 via a domain different from multimerin-2, the known ligand for CD93. In two mouse tumor models, blockade of the CD93/IGFBP7 interaction by monoclonal antibodies promoted vascular maturation to reduce leakage, leading to reduced tumor hypoxia and increased tumor perfusion. CD93 blockade in mice increased drug delivery, resulting in an improved antitumor response to gemcitabine or fluorouracil. Blockade of the CD93 pathway triggered a substantial increase in intratumoral effector T cells, thereby sensitizing mouse tumors to immune checkpoint therapy. Last, analysis of samples from patients with cancer under anti-programmed death 1/programmed death-ligand 1 treatment revealed that overexpression of the IGFBP7/CD93 pathway was associated with poor response to therapy. Thus, our study identified a molecular interaction involved in tumor vascular dysfunction and revealed an approach to promote a favorable tumor microenvironment for therapeutic intervention.


Assuntos
Neoplasias , Preparações Farmacêuticas , Animais , Células Endoteliais , Humanos , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
3.
Cell ; 176(1-2): 334-347.e12, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30580966

RESUMO

Lymphocyte-activation gene 3 (LAG-3) is an immune inhibitory receptor, with major histocompatibility complex class II (MHC-II) as a canonical ligand. However, it remains controversial whether MHC-II is solely responsible for the inhibitory function of LAG-3. Here, we demonstrate that fibrinogen-like protein 1 (FGL1), a liver-secreted protein, is a major LAG-3 functional ligand independent from MHC-II. FGL1 inhibits antigen-specific T cell activation, and ablation of FGL1 in mice promotes T cell immunity. Blockade of the FGL1-LAG-3 interaction by monoclonal antibodies stimulates tumor immunity and is therapeutic against established mouse tumors in a receptor-ligand inter-dependent manner. FGL1 is highly produced by human cancer cells, and elevated FGL1 in the plasma of cancer patients is associated with a poor prognosis and resistance to anti-PD-1/B7-H1 therapy. Our findings reveal an immune evasion mechanism and have implications for the design of cancer immunotherapy.


Assuntos
Antígenos CD/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Animais , Antígenos CD/imunologia , Linhagem Celular , Fibrinogênio/imunologia , Fibrinogênio/metabolismo , Genes MHC da Classe II/genética , Genes MHC da Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunoterapia , Ligantes , Fígado/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Neoplasias/genética , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
4.
Sci Adv ; 2(4): e1500637, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27152329

RESUMO

The central nervous system (CNS) is an immune-privileged organ with the capacity to prevent excessive inflammation. Aside from the blood-brain barrier, active immunosuppressive mechanisms remain largely unknown. We report that a neuron-specific molecule, synaptic adhesion-like molecule 5 (SALM5), is a crucial contributor to CNS immune privilege. We found that SALM5 suppressed lipopolysaccharide-induced inflammatory responses in the CNS and that a SALM-specific monoclonal antibody promoted inflammation in the CNS, and thereby aggravated clinical symptoms of mouse experimental autoimmune encephalomyelitis. In addition, we identified herpes virus entry mediator as a functional receptor that mediates SALM5's suppressive function. Our findings reveal a molecular link between the neuronal system and the immune system, and provide potential therapeutic targets for the control of CNS diseases.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Sistema Nervoso Central/imunologia , Inflamação/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Moléculas de Adesão Celular Neuronais/imunologia , Sistema Nervoso Central/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia
5.
Blood ; 115(10): 1941-8, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20068221

RESUMO

Antigen-specific memory T cells (Tms) are essential in the immune surveillance of residual and metastatic tumors. Activation of Tms requires designing vaccines based on tumor rejection antigens, which are often not available to cancer patients. Therefore, it is desirable to have a general applicable approach to activate Tms without extensive knowledge of tumor antigens. Here, we report that activation of antigen-specific Tms could be achieved by the administration of agonistic anti-CD137 monoclonal antibody without additional tumor vaccination, leading to the prevention of recurrence and metastases after surgical resection of primary tumors in mouse models. By reconstitution with CD137-deficient Tms, we demonstrate that expression of CD137 on antigen-specific Tms is only partially required for the effect of anti-CD137 antibody. Other host cells, including those from hematopoietic and nonhematopoietic origins, are also important because ablation of CD137 from these cells partially but significantly eliminates antitumor effect of anti-CD137 antibody. Our findings implicate a potential new approach to prevent recurrence and metastases in cancer patients.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias/prevenção & controle , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Animais , Vacinas Anticâncer/uso terapêutico , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/metabolismo , Memória Imunológica/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Metástase Neoplásica , Neoplasias/imunologia , Neoplasias/mortalidade , Neoplasias/patologia , Prevenção Secundária , Análise de Sobrevida , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
6.
Blood ; 113(23): 5811-8, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19339692

RESUMO

Programmed death one (PD-1) is an inducible molecule belonging to the immunoglobulin superfamily. It is expressed on activated T and B lymphocytes and plays pivotal roles in the negative regulation of adaptive immune responses. We report here an unexpected finding: that PD-1 could also be induced on splenic dendritic cells (DCs) by various inflammatory stimuli. Adoptive transfer of PD-1-deficient DCs demonstrates their superior capacity to wild-type DCs in innate protection of mice against lethal infection by Listeria monocytogenes. Furthermore, PD-1-deficient mice are also more resistant to the infection than wild-type controls, even in the absence of T and B cells, accompanied by elevated production of DC-derived interleukin-12 and tumor necrosis factor-alpha. Our results reveal a novel role of PD-1 in the negative regulation of DC function during innate immune response.


Assuntos
Antígenos de Diferenciação/imunologia , Células Dendríticas/imunologia , Imunidade Inata/imunologia , Listeriose/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Linhagem Celular , Interleucina-12/imunologia , Ligantes , Listeria monocytogenes/imunologia , Camundongos , Camundongos Knockout , Receptor de Morte Celular Programada 1 , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/imunologia
7.
Blood ; 113(8): 1759-67, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19109567

RESUMO

B7-H4 is an immunoglobulin superfamily molecule and shown to be inhibitory for T-cell responses. To explore physiologic roles of B7-H4, we created B7-H4-deficient (KO) mice by genetic targeting. B7-H4KO mice are healthy and their T- and B-cell responses to polyclonal antigens are in normal range. However, B7-H4KO mice are more resistant to infection by Listeria monocytogenes than their littermates. Within 3 days after infection, bacterial colonies in livers and spleens are significantly lower than the controls, suggesting a role of B7-H4 in enhancing innate immunity. Further studies demonstrate that neutrophils increase in peripheral organs of B7-H4KO mice more so than their littermates but their bactericidal functions remain unchanged. Augmented innate resistance is completely dependent on neutrophils, even in the absence of adaptive immunity. In vitro B7-H4 inhibits the growth of bone marrow-derived neutrophil progenitors, suggesting an inhibitory function of B7-H4 in neutrophil expansion. Our results identify B7-H4 as a negative regulator of the neutrophil response to infection and provide a new target for manipulation of innate immunity.


Assuntos
Antígeno B7-1/genética , Antígeno B7-1/imunologia , Listeriose/imunologia , Neutrófilos/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/microbiologia , Células da Medula Óssea/citologia , Antígeno CD11b/metabolismo , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Neutrófilos/microbiologia , Fagocitose/imunologia , Receptores de Quimiocinas/metabolismo , Explosão Respiratória/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Inibidor 1 da Ativação de Células T com Domínio V-Set
8.
Blood ; 111(7): 3635-43, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18223165

RESUMO

B7-H1 is an immunoglobulin-like immune suppressive molecule broadly detectable on the majority of human and rodent cancers, and its functions have been attributed to delivering an inhibitory signal to its counter-receptor programmed death-1 (PD-1) on T cells. Here we report that B7-H1 on cancer cells receives a signal from PD-1 to rapidly induce resistance against T cell-mediated killing because crippling signaling capacity of B7-H1 but not PD-1 ablates this resistance. Importantly, loss of B7-H1 signaling is accompanied by increased susceptibility to immune-mediated tumoricidal activity. In addition to resistance against T-cell destruction, B7-H1+ cancer cells also become refractory to apoptosis induced by Fas ligation or the protein kinase inhibitor Staurosporine. Our study reveals a new mechanism by which cancer cells use a receptor on immune cells as a ligand to induce resistance to therapy.


Assuntos
Antígenos de Superfície/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Apoptose/imunologia , Antígeno B7-1/imunologia , Glicoproteínas de Membrana/imunologia , Neoplasias/imunologia , Peptídeos/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Animais , Apoptose/efeitos dos fármacos , Antígeno B7-H1 , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Camundongos , Camundongos Endogâmicos DBA , Neoplasias/patologia , Receptor de Morte Celular Programada 1 , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/farmacologia , Linfócitos T/patologia , Receptor fas/imunologia
9.
Cancer Res ; 67(18): 8900-5, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875732

RESUMO

B7-H4 is a recently identified B7 family member. We previously showed that ovarian tumor and associated macrophages expressed B7-H4; tumor B7-H4+ macrophages and CD4+CD25+FOXP3+ regulatory T cells (Treg cells) suppressed tumor-associated antigen-specific T-cell immunity. To determine the pathologic relationship between B7-H4, macrophages, and Treg cells in the tumor environment, in addition to Treg cell numbers, we quantified B7-H4 expression in the tumor and tumor-associated macrophages in 103 patients with ovarian carcinoma. We observed that the intensity of B7-H4 expression in macrophages was significantly correlated with Treg cell numbers in the tumor. Further, both Treg cells and macrophage B7-H4, but not tumor B7-H4, were negatively associated with patient outcome. Tumor Treg cells enabled macrophages to spontaneously produce interleukin (IL)-10 and IL-6. Tumor macrophages stimulated B7-H4 expression in an autocrine manner through IL-10 and IL-6. Our previous work showed that tumor-associated macrophages spontaneously produced chemokine CCL22 to mediate Treg cell trafficking into tumor, and Treg cells induced B7-H4 on antigen-presenting cells (APC) including macrophages. Altogether, our data support the concept that there is a mechanistic interaction between Treg cells and macrophage, and that Treg cells may convey the suppressive activity to APCs through B7-H4 induction in human ovarian cancer.


Assuntos
Antígeno B7-1/imunologia , Neoplasias Ovarianas/imunologia , Linfócitos T Reguladores/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígeno B7-1/biossíntese , Feminino , Humanos , Interleucina-10/biossíntese , Interleucina-10/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Macrófagos/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set
10.
Blood ; 109(11): 4882-9, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17244673

RESUMO

CD137 has long been recognized as a costimulatory receptor for growth and functional maturation of recently activated T cells in the presence of T-cell receptor signal. In this report, we present the fact that, in the absence of MHC and antigen, triggering of CD137 by an agonist monoclonal antibody induces vigorous growth of both CD8(+) and CD4(+) T cells with memory phenotype, whereas it does not affect naive T cells. Moreover, T cells with memory phenotype accumulate progressively in transgenic mice overexpressing CD137 ligand. CD137-mediated proliferation of memory T cells is directly through CD137 on T cells and does not require IL-15 and IFN-gamma. Our results define a new role of CD137 signal in the growth of memory T cells.


Assuntos
Memória Imunológica , Linfócitos T/imunologia , Linfócitos T/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/biossíntese , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Divisão Celular , Interferon gama/metabolismo , Interleucina-15/metabolismo , Ligantes , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
11.
Blood ; 109(9): 4097-104, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17179227

RESUMO

Decoy lymphotoxin beta receptor (LTbetaR) has potent immune inhibitory activities and thus represents a promising biologic for the treatment of inflammation, autoimmune diseases, and graft-versus-host disease (GVHD). As this reagent interrupts multiple molecular interactions, including LTbeta-LTbetaR and LIGHT-HVEM/LTbetaR, underlying molecular mechanisms have yet to be fully understood. In this study, we demonstrate that blockade of the LIGHT-HVEM pathway is sufficient to induce amelioration of GVHD in mouse models. Anti-host cytotoxic T lymphocyte (CTL) activity following in vivo transfer of allogeneic lymphocytes was completely abrogated when LIGHT- or HVEM-deficient (KO) T cells were used as donor cells. Accordingly, survival of the recipient mice following the transfer of allogeneic bone marrow cells plus LIGHT-KO or HVEM-KO T cells was significantly prolonged. In the absence of LIGHT-HVEM costimulation, alloreactive donor T cells undergo vigorous apoptosis while their proliferative potential remains intact. Furthermore, we prepared a neutralizing monoclonal antibody (mAb) specific to HVEM and showed that administration of anti-HVEM mAb profoundly ameliorated GVHD and led to complete hematopoietic chimerism with donor cells. Collectively, our results demonstrate an indispensable role of LIGHT-HVEM costimulation in the pathogenesis of GVHD and illustrate a novel target for selective immunotherapy in allogeneic bone marrow transplantation.


Assuntos
Anticorpos Monoclonais/farmacologia , Transplante de Medula Óssea , Doença Enxerto-Hospedeiro/terapia , Transfusão de Linfócitos , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Quimeras de Transplante/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/terapia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Imunoterapia , Inflamação/imunologia , Inflamação/patologia , Inflamação/terapia , Camundongos , Camundongos Endogâmicos BALB C , Transplante Homólogo
12.
J Exp Med ; 203(4): 871-81, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16606666

RESUMO

Tumor-associated macrophages are a prominent component of ovarian cancer stroma and contribute to tumor progression. B7-H4 is a recently identified B7 family molecule. We show that primary ovarian tumor cells express intracellular B7-H4, whereas a fraction of tumor macrophages expresses surface B7-H4. B7-H4+ tumor macrophages, but not primary ovarian tumor cells, suppress tumor-associated antigen-specific T cell immunity. Blocking B7-H4-, but not arginase-, inducible nitric oxide synthase or B7-H1 restored the T cell stimulating capacity of the macrophages and contributes to tumor regression in vivo. Interleukin (IL)-6 and IL-10 are found in high concentrations in the tumor microenvironment. These cytokines stimulate macrophage B7-H4 expression. In contrast, granulocyte/macrophage colony-stimulating factor and IL-4, which are limited in the tumor microenvironment, inhibit B7-H4 expression. Ectopic expression of B7-H4 makes normal macrophages suppressive. Thus, B7-H4+ tumor macrophages constitute a novel suppressor cell population in ovarian cancer. B7-H4 expression represents a critical checkpoint in determining host responses to dysfunctional cytokines in ovarian cancer. Blocking B7-H4 or depleting B7-H4+ tumor macrophages may represent novel strategies to enhance T cell tumor immunity in cancer.


Assuntos
Antígeno B7-1/genética , Carcinoma/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Antígenos CD/fisiologia , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-H1 , Biomarcadores , Carcinoma/imunologia , Feminino , Humanos , Imunofenotipagem , Interleucina-10/fisiologia , Interleucina-6/fisiologia , Macrófagos/classificação , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Ovarianas/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Células Tumorais Cultivadas , Inibidor 1 da Ativação de Células T com Domínio V-Set
13.
J Clin Invest ; 116(4): 1045-51, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16557300

RESUMO

LIGHT is an important costimulatory molecule for T cell immunity. Recent studies have further implicated its role in innate immunity and inflammatory diseases, but its cellular and molecular mechanisms remain elusive. We report here that LIGHT is upregulated and functions as a proinflammatory cytokine in 2 independent experimental hepatitis models, induced by concanavalin A and Listeria monocytogenes. Molecular mutagenesis studies suggest that soluble LIGHT protein produced by cleavage from the cell membrane plays an important role in this effect through the interaction with the lymphotoxin-beta receptor (LTbetaR) but not herpes virus entry mediator. NK1.1+ T cells contribute to the production, but not the cleavage or effector functions, of soluble LIGHT. Importantly, treatment with a mAb that specifically interferes with the LIGHT-LTbetaR interaction protects mice from lethal hepatitis. Our studies thus identify a what we believe to be a novel function of soluble LIGHT in vivo and offer a potential target for therapeutic interventions in hepatic inflammatory diseases.


Assuntos
Citocinas/metabolismo , Hepatite/etiologia , Proteínas de Membrana/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Antígenos Ly , Antígenos de Superfície/metabolismo , Concanavalina A/metabolismo , Concanavalina A/farmacologia , Hepatite/metabolismo , Inflamação/metabolismo , Lectinas Tipo C/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Receptor beta de Linfotoxina , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral , Receptores Virais/imunologia , Receptores Virais/metabolismo , Solubilidade , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
14.
FEBS Lett ; 579(27): 6259-64, 2005 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16253242

RESUMO

The co-signaling molecule B7-H1 (CD274) functions as both a co-inhibitor through programmed death-1 (PD-1) receptor and a co-stimulator via an as-yet-unidentified receptor on T cells. We investigated the physiological role of endogenous B7-H1 in the pathogenesis of herpetic stromal keratitis (HSK) caused by herpes simplex virus type 1 (HSV-1). Following HSV-1 infection of the cornea of mice, B7-H1 expression was up-regulated in the CD11b+ macrophage population in the draining lymph nodes (dLN) and in the inflamed cornea. In addition, HSV-1 infection significantly increased PD-1 expression on CD4+ T cells in the dLN and inflamed cornea. The administration of antagonistic B7-H1 monoclonal antibody resulted in the proliferation of HSV-specific CD4+ T cells that secreted interferon (INF)-gamma, and inhibited the apoptosis of HSV-specific CD4+ T cells, which exaggerated HSK. These results strongly suggest that the B7-H1 may be involved in suppression of the development of HSK.


Assuntos
Antígeno B7-1/fisiologia , Linfócitos T CD4-Positivos/imunologia , Herpesvirus Humano 1 , Ceratite Herpética/imunologia , Glicoproteínas de Membrana/fisiologia , Peptídeos/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Antígeno B7-H1 , Antígeno CD11b/análise , Córnea/imunologia , Córnea/patologia , Interferon gama/metabolismo , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/antagonistas & inibidores , Células Estromais/metabolismo , Células Estromais/virologia , Células Th1/imunologia , Regulação para Cima
15.
Cancer Res ; 65(3): 1089-96, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15705911

RESUMO

Contemporary approaches for vaccination and immunotherapy are often capable of eliciting strong T-cell responses against tumor antigens. However, such responses are not parallel to clinical tumor regression. The development of evasion mechanisms within tumor microenvironment may be responsible for poor therapeutic responses. We report here that constitutive or inducible expression of B7-H1, a B7 family molecule widely expressed by cancers, confers resistance to therapeutic anti-CD137 antibody in mice with established tumors. The resistance is accompanied with failure of antigen-specific CD8+ CTLs to destroy tumor cells without impairment of CTL function. Blockade of B7-H1 or PD-1 by specific monoclonal antibodies could reverse this resistance and profoundly enhance therapeutic efficacy. Our findings support that B7-H1/PD-1 forms a molecular shield to prevent destruction by CTLs and implicate new approaches for immunotherapy of human cancers.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Superfície/imunologia , Antígeno B7-1/imunologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Neoplasias Experimentais/imunologia , Peptídeos/antagonistas & inibidores , Peptídeos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Proteínas Reguladoras de Apoptose , Antígeno B7-1/biossíntese , Antígeno B7-H1 , Anergia Clonal , Feminino , Imunoterapia/métodos , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1 , Linfócitos T Citotóxicos/imunologia
16.
J Immunol ; 173(9): 5445-50, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15494491

RESUMO

B7-H3 is a B7 family molecule with T cell costimulatory function in vitro. The in vivo role of B7-H3 in the stimulation of tumor immunity is unclear. We report here that expression of B7-H3 by transfection of the mouse P815 tumor line enhances its immunogenicity, leading to the regression of tumors and amplification of a tumor-specific CD8+ CTL response in syngeneic mice. Tumor cells engineered to express B7-H3 elicit a rapid clonal expansion of P1A tumor Ag-specific CD8+ CTL in lymphoid organs in vivo and acquire the ability to directly stimulate T cell growth, division, and development of cytolytic activity in vitro. Our results thus establish a role for B7-H3 in the costimulation of T cell immune responses in vivo.


Assuntos
Adjuvantes Imunológicos/fisiologia , Antígeno B7-1/fisiologia , Epitopos de Linfócito T/imunologia , Ativação Linfocitária/imunologia , Sarcoma de Mastócitos/imunologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos B7 , Antígeno B7-1/genética , Células CHO , Divisão Celular/imunologia , Linhagem Celular Tumoral , Células Clonais , Cricetinae , Testes Imunológicos de Citotoxicidade , DNA Complementar/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Nus , Camundongos Transgênicos , Transfecção
17.
Immunity ; 20(3): 327-36, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15030776

RESUMO

Upon systemic activation by antigens, CD8(+), but not CD4(+), T cells selectively accumulate and undergo apoptosis in the liver, a mechanism associated with the induction of hepatic tolerance and chronic infection. The molecular basis for CD8(+) T cell preference in this process is unknown. We prepared B7-H1-deficient mice by gene targeting and found spontaneous accumulation of CD8(+) T cells in the liver while CD4(+) T cell levels remained normal. Moreover, antigen-driven CD8(+) T cells proliferated normally while apoptotic levels during the contraction phase was selectively impaired in the liver, leading to accelerated hepatocyte damage in experimental autoimmune hepatitis. Therefore, B7-H1 is a key protein selectively regulating the accumulation and deletion of intrahepatic CD8(+) T cells and may also contribute to inflammation, autoimmune diseases, and tolerance in the liver.


Assuntos
Apoptose , Antígeno B7-1/fisiologia , Proteínas Sanguíneas , Linfócitos T CD8-Positivos/imunologia , Fígado/imunologia , Peptídeos , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Antígeno B7-1/genética , Antígeno B7-H1 , Linfócitos T CD8-Positivos/patologia , Movimento Celular , Hepatite Animal/imunologia , Hepatite Animal/patologia , Cinética , Fígado/citologia , Fígado/patologia , Ativação Linfocitária , Glicoproteínas de Membrana , Camundongos , Camundongos Knockout
18.
Blood ; 103(1): 177-84, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12969968

RESUMO

T-cell anergy is a tolerance mechanism defined as a hyporesponsive status of antigen-specific T cells upon prior antigen encounter and is believed to play a critical role in the evasion of tumor immunity and the amelioration of allogeneic transplant rejection. Molecular mechanisms in controlling T-cell anergy are less known. We show here that administration of an agonistic monoclonal antibody (mAb) to CD137, a member of the tumor necrosis factor receptor superfamily, prevents the induction of CD8+ cytolytic T-lymphocyte (CTL) anergy by soluble antigens. More importantly, CD137 mAb restores the functions of established anergic CTLs upon reencountering their cognate antigen. As a result, infusion of CD137 mAb inhibits progressive tumor growth that is caused by soluble tumor antigen-induced tolerance in a P815R model. CD137 mAb also restores proliferation and effector functions of anergic alloreactive 2C T cells in a bone marrow transplantation model. Our results indicate that ligation of CD137 receptor delivers a regulatory signal for T-cell anergy and implicate manipulation of the CD137 pathway as a new approach to break T-cell tolerance.


Assuntos
Anergia Clonal , Receptores de Fator de Crescimento Neural/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos CD , Transplante de Medula Óssea , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Ovalbumina/imunologia , Quimera por Radiação/imunologia , Receptores de Fator de Crescimento Neural/agonistas , Receptores de Fator de Crescimento Neural/antagonistas & inibidores , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Transdução de Sinais , Transplante Homólogo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral
19.
J Immunol ; 171(9): 4650-4, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14568939

RESUMO

B7-H4 is a recently identified B7 family member that negatively regulates T cell immunity by the inhibition of T cell proliferation, cytokine production, and cell cycle progression. In this study, we report that the genomic DNA of human B7-H4 is mapped on chromosome 1 comprised of six exons and five introns spanning 66 kb, of which exon 6 is used for alternative splicing to generate two different transcripts. Similar B7-H4 structure is also found in mouse genomic DNA in chromosome 3. A human B7-H4 pseudogene is identified in chromosome 20p11.1 with a single exon and two stop codons in the coding region. Immunohistochemistry analysis using B7-H4-specific mAb demonstrates that B7-H4 is not expressed on the majority of normal human tissues. In contrast, up to 85% (22 of 26) of ovarian cancer and 31% (5 of 16) of lung cancer tissues constitutively express B7-H4. Our results indicate a tight regulation of B7-H4 expression in the translational level in normal peripheral tissues and a potential role of B7-H4 in the evasion of tumor immunity.


Assuntos
Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Regulação para Baixo/imunologia , Perfilação da Expressão Gênica , Família Multigênica/imunologia , Análise de Sequência de DNA , Animais , Antígeno B7-1/química , Linhagem Celular Tumoral , Regulação para Baixo/genética , Éxons/genética , Feminino , Perfilação da Expressão Gênica/métodos , Ordem dos Genes/imunologia , Humanos , Íntrons/genética , Neoplasias Pulmonares/química , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Neoplasias Ovarianas/química , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Análise de Sequência de DNA/métodos , Transcrição Gênica/imunologia , Inibidor 1 da Ativação de Células T com Domínio V-Set
20.
Immunity ; 18(6): 849-61, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12818165

RESUMO

We identify a B7 family molecule, B7-H4, by protein sequence analysis and comparative molecular modeling. While B7-H4 mRNA is widely distributed in mouse and human peripheral tissues, cell surface expression of B7-H4 protein is limited and shows an inducible pattern on hematopoietic cells. Putative receptor of B7-H4 can be upregulated on activated T cells. By arresting cell cycle, B7-H4 ligation of T cells has a profound inhibitory effect on the growth, cytokine secretion, and development of cytotoxicity. Administration of B7-H4Ig into mice impairs antigen-specific T cell responses whereas blockade of endogenous B7-H4 by specific monoclonal antibody promotes T cell responses. B7-H4 thus may participate in negative regulation of cell-mediated immunity in peripheral tissues.


Assuntos
Antígeno B7-1/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Sequência de Aminoácidos , Animais , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Northern Blotting , Ciclo Celular/fisiologia , Perfilação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Inibidor 1 da Ativação de Células T com Domínio V-Set
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA