Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Cancer ; 137(5): 1085-94, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25683463

RESUMO

Uveal melanoma (UM) is the most common intraocular tumor in adults and liver metastasis is the leading cause of death in UM patients. We have previously shown that NKT cell-deficient mice develop significantly fewer liver metastases from intraocular melanomas than do wild-type (WT) mice. Here, we examine the interplay between liver NKT cells and NK cells in resistance to liver metastases from intraocular melanomas. NKT cell-deficient CD1d(-/-) mice and WT C57BL/6 mice treated with anti-CD1d antibody developed significantly fewer liver metastases than WT mice following either intraocular or intrasplenic injection of B16LS9 melanoma cells. The increased number of metastases in WT mice was associated with reduced liver NK cytotoxicity and decreased production of IFN-γ. However, liver NK cell-mediated cytotoxic activity was identical in non-tumor bearing NKT cell-deficient mice and WT mice, indicating that liver metastases were crucial for the suppression of liver NK cells. Depressed liver NK cytotoxicity in WT mice was associated with production of IL-10 by bone marrow-derived liver cells that were neither Kupffer cells nor myeloid-derived suppressor cells and by increased IL-10 receptor expression on liver NK cells. IL-10(-/-) mice had significantly fewer liver metastases than WT mice, but were not significantly different from NKT cell-deficient mice. Thus, development of melanoma liver metastases is associated with upregulation of IL-10 in the liver and an elevated expression of IL-10 receptor on liver NK cells. This impairment of liver NK activity is NKT cell-dependent and only occurs in hosts with melanoma liver metastases.


Assuntos
Antígenos CD1d/metabolismo , Células Matadoras Naturais/fisiologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Melanoma/imunologia , Células T Matadoras Naturais/fisiologia , Neoplasias Uveais/imunologia , Animais , Anticorpos/farmacologia , Antígenos CD1d/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Interferon gama/metabolismo , Interleucina-10/metabolismo , Neoplasias Hepáticas/patologia , Ativação Linfocitária , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Receptores de Interleucina-10/metabolismo , Neoplasias Uveais/patologia
2.
J Immunol ; 187(8): 4219-28, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21918192

RESUMO

Although intraocular tumors reside in an immune-privileged site where immune responses are suppressed, some tumors are rejected. An example of this is the rejection of intraocular adenovirus-induced (adenovirus type 5 early region 1 [Ad5E1]) tumors in C57BL/6 mice. We previously identified an Ad5E1 tumor clone in which the rejection is IFN-γ dependent and culminates in the destruction of both the tumor and the eye. Although Ad5E1 tumors are not rejected when transplanted into the eyes of IFN-γ KO mice, they are rejected after s.c. transplantation. Thus, outside of the eye Ad5E1 tumors elicit a form of tumor immunity that is IFN-γ independent. In this article, we demonstrate that IFN-γ-independent s.c. rejection requires both CD4(+) and CD8(+) T cells. Furthermore, s.c. tumor rejection requires IL-17, which is produced by IFN-γ-deficient CD4(+) T cells in response to tumor Ags (TAs). Splenocytes from CD4-depleted IFN-γ KO mice produce significantly less IL-17 compared with splenocytes from isotype-treated IFN-γ KO animals in response to TAs. Furthermore, depletion of IL-17 decreases CTL activity against Ad5E1 tumor cells. In this model we propose that, in the absence of IFN-γ, CD4(+) T cells produce IL-17 in response to TAs, which increases CTL activity that mediates tumor rejection; however, this does not occur in the eye. IL-6 production within the eye is severely reduced, which is consistent with the failure to induce Th17 cells within the intraocular tumors. In contrast, the s.c. environment is replete with IL-6 and supports the induction of Th17 cells. Therefore, IFN-γ-independent tumor rejection is excluded from the eye and may represent a newly recognized form of ocular immune privilege.


Assuntos
Olho/imunologia , Interferon gama/imunologia , Interleucina-17/imunologia , Neoplasias Experimentais/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
J Immunol ; 186(12): 6737-45, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21551366

RESUMO

IL-17A is a proinflammatory cytokine that has received attention for its role in the pathogenesis of several autoimmune diseases. IL-17A has also been implicated in cardiac and renal allograft rejection. Accordingly, we hypothesized that depletion of IL-17A would enhance corneal allograft survival. Instead, our results demonstrate that blocking IL-17A in a mouse model of keratoplasty accelerated the tempo and increased the incidence of allograft rejection from 50 to 90%. We describe a novel mechanism by which CD4(+)CD25(+) regulatory T cells (Tregs) respond to IL-17A and enhance corneal allograft survival. Our findings suggest the following: 1) IL-17A is necessary for ocular immune privilege; 2) IL-17A is not required for the induction of anterior chamber-associated immune deviation; 3) Tregs require IL-17A to mediate a contact-dependent suppression; 4) corneal allograft Tregs suppress the efferent arm of the immune response and are Ag specific; 5) Tregs are not required for corneal allograft survival beyond day 30; and 6) corneal allograft-induced Treg-mediated suppression is transient. Our findings identify IL-17A as a cytokine essential for the maintenance of corneal immune privilege and establish a new paradigm whereby interplay between IL-17A and CD4(+)CD25(+) Tregs is necessary for survival of corneal allografts.


Assuntos
Transplante de Córnea/métodos , Sobrevivência de Enxerto/imunologia , Imunossupressores , Interleucina-17/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos , Rejeição de Enxerto/imunologia , Fenômenos do Sistema Imunitário , Interleucina-17/farmacologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos
4.
J Immunol ; 185(8): 4651-8, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844197

RESUMO

Corneal allograft rejection has been described as a Th1-mediated process involving IFN-γ production. However, it has been reported that corneal allograft rejection soars in IFN-γ(-/-) mice or mice treated with anti-IFN-γ mAb. Th17 is a recently described IL-17A-producing Th cell population that has been linked to renal and cardiac graft rejection, which was originally thought to be Th1-mediated. We tested the hypothesis that Th17 cells mediate corneal allograft rejection in an IL-17A-dependent fashion and unexpectedly found that depletion of IL-17A increased the incidence of rejection to 90%. We demonstrate that the exacerbated rejection following depletion of IL-17A did not result from a loss of cross-regulation of Th1 cells or exaggerated delayed-type hypersensitivity responses. Instead, inhibition of the Th1 or Th17 cell lineages promoted the emergence of a Th2 cell subset that independently mediated allograft rejection. These findings demonstrate that IL-17A is not required for corneal allograft rejection and may instead contribute to the immune privilege of corneal allografts.


Assuntos
Transplante de Córnea , Rejeição de Enxerto/imunologia , Interleucina-17/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Imunologia de Transplantes , Transferência Adotiva , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Sobrevivência de Enxerto , Imuno-Histoquímica , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante Homólogo
5.
J Immunol ; 184(11): 6076-83, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20410484

RESUMO

Allergic conjunctivitis (AC) and airway hyperreactivity exacerbate corneal allograft rejection. Because AC and airway hyperreactivity are allergic diseases of mucosal tissues, we determined whether an allergic disease of a nonmucosal tissue would affect corneal allograft rejection and whether Th2 cells alone accounted for accelerated graft rejection in allergic mice. Hosts sensitized cutaneously with short ragweed pollen developed cutaneous immediate hypersensitivity but rejected corneal allografts at the same tempo and incidence as naive mice. Th2 immune deviation induced with keyhole limpet hemocyanin and IFA did not affect corneal allograft rejection. Thus, Th2 immune deviation alone does not account for the exacerbation of corneal allograft rejection that occurs in mice with AC. CD4(+) T cells from AC mice elaborated Th1 (IFN-gamma) and Th2 (IL-13) cytokines when challenged with donor alloantigens. Adoptive transfer of Th1 or Th2 cells to nude mice, from AC mice that had rejected corneal allografts, produced graft rejection in 70% and 20% of the hosts, respectively. In contrast, adoptive transfer of a combination of Th1 and Th2 cells produced 100% rejection. Administration of exogenous IFN-gamma could substitute for Th1 cells and produced 100% corneal allograft rejection in recipients of Th2 cells alone. By contrast, IFN-gamma did not significantly enhance corneal allograft rejection mediated by Th1 cells. Thus, exacerbation of corneal allograft rejection in mice with AC is associated with a mixed Th1 and Th2 alloimmune response, and the contribution of Th1 cells is through their production of IFN-gamma.


Assuntos
Conjuntivite Alérgica/imunologia , Rejeição de Enxerto/imunologia , Ativação Linfocitária/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Separação Celular , Conjuntivite Alérgica/complicações , Transplante de Córnea , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Rejeição de Enxerto/complicações , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante Homólogo
6.
Int Immunol ; 22(8): 627-36, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20504886

RESUMO

Allergic conjunctivitis (AC) is elicited by immediate hypersensitivity responses to environmental agents. It is initiated by a T(h)2-dominated immune response that is characterized by production of IgE antibodies and eosinophilic infiltration. By using an experimental mouse model of AC induced by short ragweed (SRW) pollen, we show that sensitized Jalpha18(-/-) mice, which lack type I NKT cells, and CD1d(-/-) mice, which lack type I and type II NKT cells, exhibited a decrease in tearing, lid edema, conjunctival edema and vasodilatation and eosinophil infiltration into the conjunctiva when compared with wild-type (WT) mice in both T(h)1- and T(h)2-prone hosts (C57BL/6 and BALB/c mice, respectively). This demonstrates that NKT cells are needed for both the early and late phases of AC. Adoptive transfer of SRW-primed CD4(+) T cells from Jalpha18(-/-) mice into naive WT BALB/c mice revealed that NKT cells were needed for the maximal induction of allergen-specific T(h)2 cells. Results from adoptive transfer of SRW-primed CD4(+) T cells from WT BALB/c mice to naive Jalpha18(-/-) mice indicated that NKT cells were also needed for the expression of AC produced by allergen-primed CD4(+) T cells. The decreased expression of AC in NKT cell-deficient mice was correlated with significant reduction in the production of T(h)2 cytokines in SRW pollen-sensitized mice compared with WT mice and in the capacity of SRW pollen-sensitized CD4(+) T cells to mediate ocular inflammation when the hosts were confronted with SRW pollen at the ocular surface.


Assuntos
Conjuntivite Alérgica/imunologia , Células T Matadoras Naturais/imunologia , Animais , Conjuntivite Alérgica/metabolismo , Conjuntivite Alérgica/fisiopatologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th2/imunologia
7.
Immunology ; 123(3): 367-77, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17944931

RESUMO

Although intraocular tumours reside in an immune-privileged site, they can circumvent immune privilege and undergo rejection. Ocular tumour rejection typically follows one of two pathways. One pathway involves CD4+ T cells, delayed-type hypersensitivity (DTH), and culmination in ischemic necrosis of the tumour and phthisis (atrophy) of the eye. The second pathway is DTH-independent and does not inflict collateral injury to ocular tissues, and the eye is preserved. In this study, we used a well-characterized tumour, Ad5E1, to investigate the role of CD4+ T cells in the non-phthisical form of intraocular tumour rejection. It has been previously documented that CD4+ T cells and interferon (IFN)-gamma are necessary for rejection of these tumours in the eye. In this study, we demonstrate that CD4+ T cells can circumvent immune privilege and infiltrate intraocular Ad5E1 tumours. Following tumour rejection, CD4+ T cells from tumour rejector mice could be adoptively transferred to severe combined immunodeficiency (SCID) mice and protect them from intraocular Ad5E1 tumour growth. Tumour-specific CD4+ T cells produced IFN-gamma in response to Ad5E1 tumour antigens. Macrophages also contributed to rejection, as they were present in intraocular Ad5E1 tumours, and local depletion of macrophages resulted in progressive tumour growth. Ocular macrophages contributed to Ad5E1 tumour rejection, as Ad5E1 tumour rejection did not occur in macrophage-depleted SCID mice reconstituted with rejector CD4+ T cells. This demonstrates that macrophage and CD4+ T-cell co-operation is needed for non-phthisical rejection of intraocular tumours.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Neoplasias Oculares/imunologia , Macrófagos/imunologia , Transferência Adotiva , Animais , Células Cultivadas , Citotoxicidade Imunológica/imunologia , Progressão da Doença , Ensaio de Imunoadsorção Enzimática/métodos , Neoplasias Oculares/patologia , Neoplasias Oculares/prevenção & controle , Hipersensibilidade Tardia/imunologia , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias
8.
Invest Ophthalmol Vis Sci ; 49(6): 2518-25, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18296654

RESUMO

PURPOSE: To assess the expression of PD-L1 on human uveal melanomas and its potential to suppress T-cell function. METHODS: A panel of primary and metastatic uveal melanoma cell lines was evaluated for PD-L1 expression by RT-PCR and flow cytometric analysis. Uveal melanoma-containing eyes were examined for PD-L1 expression by immunohistochemistry. PD-L1 function was tested by coculturing IFN-gamma-pretreated uveal melanoma cells with activated Jurkat T cells for 48 hours and assessing T-cell production of IL-2 by ELISA. RESULTS: Five of the nine primary and one of the five metastatic uveal melanoma cell lines tested constitutively expressed PD-L1 protein at various levels. However, all primary and metastatic uveal melanoma cell lines upregulated PD-L1 expression after stimulation with IFN-gamma. Immunohistochemistry demonstrated that PD-L1 was not expressed by primary uveal melanomas in situ. IL-2 production by activated Jurkat T cells was decreased significantly when the cells were cocultured with IFN-gamma-pretreated uveal melanoma cells. More than 70% of IL-2 production was restored by addition of either anti-PD-L1 or anti-PD-1 antibody to the coculture assays (P < 0.01). CONCLUSIONS: Expression of PD-L1 by uveal melanoma cells regulates T-cell function by suppressing IL-2 production. The results imply that the presence of IFN-gamma in the tumor local microenvironment promotes upregulation of PD-L1 expression by uveal melanoma, which may, in part, promote immune escape by impairing T-cell function. The selective blockade of PD-L1 is a potential strategy in T-cell-based immunotherapy for uveal melanoma.


Assuntos
Antígenos CD/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Melanoma/imunologia , Proteínas de Neoplasias/metabolismo , Linfócitos T/fisiologia , Neoplasias Uveais/imunologia , Antígenos CD/genética , Proteínas Reguladoras de Apoptose/genética , Antígeno B7-H1 , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas Imunoenzimáticas , Interferon gama/farmacologia , Interleucina-2/metabolismo , Células Jurkat , Ativação Linfocitária , Melanoma/patologia , Metástase Neoplásica , Receptor de Morte Celular Programada 1 , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Uveais/patologia
9.
J Leukoc Biol ; 81(2): 421-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17077163

RESUMO

Although intraocular tumors reside in an immune-privileged site, they can circumvent immune privilege and undergo rejection, which typically follows one of two pathways. One pathway involves CD4(+) T cells, delayed-type hypersensitivity (DTH), and the culmination in ischemic necrosis of the tumor and phthisis (atrophy) of the eye. The second pathway is DTH-independent and does not inflict collateral injury to ocular tissues, and the eye is preserved. In this study, we used a well-characterized tumor, Ad5E1, to analyze the role of IFN-gamma in the nonphthisical form of intraocular tumor rejection. The results showed that IFN-gamma induced tumor cell apoptosis, inhibited tumor cell proliferation, and promoted rejection by inhibiting angiogenesis. Microarray analysis revealed that IFN-gamma induced up-regulation of five antiangiogenic genes and down-regulation of four proangiogenic genes in Ad5E1 tumor cells. Although IFN-gamma knockout (KO) mice have progressively growing intraocular tumors, IFN-gamma was not needed for the elimination of extraocular tumors, as all IFN-gamma KO mice rejected s.c. tumor inocula. This represents a heretofore unrecognized role for IFN-gamma in circumventing ocular immune privilege and eliminating intraocular tumors. The findings also reveal that some IFN-gamma-independent tumor rejection processes are excluded from the eye and may represent a new facet of ocular immune privilege.


Assuntos
Câmara Anterior/imunologia , Linfócitos T CD4-Positivos/imunologia , Neoplasias Oculares/imunologia , Rejeição de Enxerto/imunologia , Interferon gama/imunologia , Animais , Câmara Anterior/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Neoplasias Oculares/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante de Neoplasias/imunologia , Neovascularização Patológica/genética , Microglobulina beta-2/imunologia
10.
Chem Immunol Allergy ; 92: 276-289, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17264503

RESUMO

The immune surveillance theory proposed almost half a century ago stated that the immune system was responsible for preventing the formation of spontaneous tumors by identifying and eliminating neoplastic cells early in their development. Recent studies demonstrating that innate and adaptive immune effector cells participate in preventing tumor growth and are effective in reducing the frequency of tumors have revived interest in immune surveillance. Paradoxically, other recent studies demonstrate that the immune system can also promote tumor progression by altering the immunogenic phenotype of developing tumors in a process called immunoediting. These data raise new questions regarding whether immune surveillance and immunoediting occur within the immune-privileged ocular environment where the innate and adaptive immune effector cells are inhibited and/or participate in the development of regulatory T cells.


Assuntos
Neoplasias Oculares/etiologia , Neoplasias Oculares/imunologia , Animais , Olho/imunologia , Humanos , Imunidade Inata , Interferon gama/fisiologia , Células Matadoras Naturais/imunologia , Evasão Tumoral
11.
Curr Eye Res ; 31(1): 43-55, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16421019

RESUMO

Ocular immune privilege preserves the visual axis by preventing the induction of sight-threatening nonspecific inflammation. Although privilege is essential for maintaining visual integrity, intraocular tumors exploit the privileged environment and grow progressively within the anterior chamber of the eye. Recently, a large number of laboratories have constructed genetically engineered tumor cell vaccines that express high levels of costimulatory signals. These vaccines are designed to bypass the normal pathways of T-cell activation and directly activate CD8+ tumor-specific T cells. In the following series of experiments, we determined whether a tumor cell vaccine that uses costimulatory signals (CD80 and IL-12) is capable of eliminating tumors within the immune-privileged anterior chamber. As expected, vaccine-immunized mice rejected subcutaneous flank tumors (a non-privileged site). However, the vaccine failed to protect mice from even a small number of tumor cells transplanted into the immune-privileged anterior chamber. Surprisingly, immunized mice that were simultaneously challenged with subcutaneous and anterior chamber tumors were unable to eliminate tumors at either site. The failure of systemic protective immunity coincided with the loss of tumor-specific delayed hypersensitivity and cytotoxic T cells. We conclude that tumor cell vaccines that induce complete protection against tumors in non-immune-privileged sites fail to protect against the same tumor within an ocular immune-privileged site. Moreover, a tumor that escapes elimination within the eye can terminate systemic protective immunity that is induced by the tumor cell vaccine.


Assuntos
Câmara Anterior/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Neoplasias Oculares/imunologia , Linfócitos do Interstício Tumoral/imunologia , Mastocitoma/imunologia , Animais , Antígenos de Neoplasias/imunologia , Antígeno B7-1/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Neoplasias Oculares/prevenção & controle , Feminino , Hipersensibilidade Tardia/imunologia , Imunidade Celular , Imunização , Interleucina-12/imunologia , Teste de Cultura Mista de Linfócitos , Mastocitoma/prevenção & controle , Camundongos , Camundongos Endogâmicos DBA , Camundongos SCID , Transplante de Neoplasias , Linfócitos T Citotóxicos/imunologia
12.
Cancer Res ; 64(5): 1867-74, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14996751

RESUMO

Mouse tumor cells transfected with syngeneic MHC class II and costimulatory molecule genes are therapeutic vaccines in mice, provided they do not coexpress the class II-associated invariant chain (Ii). We demonstrated previously that the vaccine cells present tumor peptides via the endogenous antigen presentation pathway to activate CD4(+) and CD8(+) T cells. Because of their efficacy in mice, we are translating this vaccine strategy for clinical use. To obtain MHC class II(+)CD80(+)Ii(-) human tumor cells, we developed retroviruses encoding HLA-DR and CD80. The HLA-DR virus encodes the DRalpha and DRbeta0101 chains using an internal ribosomal entry site to coordinate expression. SUM159PT mammary carcinoma and Mel 202 ocular melanoma cells transduced with the retroviruses DRB1/CD80 express high levels of DRB0101 and CD80 on the cell surface in the absence of Ii. Irradiated SUM159PT/DR1/CD80 vaccines stimulate proliferation of non-HLA-DRB0101 peripheral blood mononuclear cells and present an exogenous DR1-restricted tetanus toxoid (TT) peptide, indicating that the transduced DRB0101 is functional. SUM159PT/DR1/CD80 vaccines were further transduced with a retrovirus encoding the TT fragment C gene, as a model tumor antigen. These cells stimulate IFN-gamma release from TT-primed human DRB0101 peripheral blood mononuclear cells, demonstrating their ability to present "endogenous" tumor antigen. Depletion and antibody blocking experiments confirm that MHC class II-restricted, endogenously synthesized epitopes are presented to CD4(+) T cells. Therefore, the MHC class II vaccines are efficient antigen-presenting cells that activate tumor-specific MHC class II-restricted, CD4(+) T lymphocytes, and they are a novel and potential immunotherapeutic for metastatic cancers.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/imunologia , Antígenos HLA-DR/genética , Ativação Linfocitária , Sequência de Aminoácidos , Antígeno B7-1/genética , Humanos , Imunoterapia , Dados de Sequência Molecular , Retroviridae/genética , Toxoide Tetânico/imunologia , Transdução Genética
13.
Invest Ophthalmol Vis Sci ; 54(1): 234-43, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23188729

RESUMO

PURPOSE: Expression of the chemokine receptor CXCR4 by tumors is associated with metastatic migration and invasion of tumor cells. The importance of CXCR4 expression by uveal melanomas in metastasis to the liver was recently demonstrated when injection of CXCR4-negative uveal melanoma cells into mice resulted in reduced liver metastasis compared with CXCR4-positive uveal melanoma cells. Factors in the eye can induce downregulation of genes by epigenetic mechanisms. This study examined whether epigenetic regulation by the ocular environment induced downregulation of CXCR4 expression. METHODS: LS174T colon cancer cells were injected in the anterior chamber (AC), subcutaneously (SC), or in the spleen capsule to induce liver metastasis in immune-deficient mice. CXCR4 gene transcription was analyzed by RT-PCR, and protein expression was determined by flow cytometry. Methyltransferase and histone deacetylase activities were determined by ELISA. Treatment with either 5-Aza-2-deoxycytidine (5-Aza) or trichostatin A (TSA) was used to induce demethylation or inhibit histone deacetylases, respectively. RESULTS: AC-derived LS174T cells showed lower CXCR4 gene expression compared with SC-, liver-derived, or wild-type tumor cells. AC-derived LS174T tumor cells expressed methyltransferase activity compared with SC-, liver-derived, and wild-type tumor cells. Deacetylase activity was elevated in AC-derived LS174T tumor cells compared with SC-derived, liver-derived, and wild-type tumor cells. Treatment of AC-derived LS174T tumor cells with 5-Aza upregulated CXCR4 expression. TSA treatment did not restore CXCR4 expression. CONCLUSIONS: These studies demonstrate that ocular microenvironment factors induce methylation and downregulation of tumor CXCR4 expression.


Assuntos
Câmara Anterior/metabolismo , Epigênese Genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Animais , Câmara Anterior/patologia , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Ceco/metabolismo , Ceco/patologia , Linhagem Celular Tumoral , Decitabina , Regulação para Baixo , Citometria de Fluxo , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/fisiologia , Inibidores de Histona Desacetilases/administração & dosagem , Histona Desacetilases/metabolismo , Histonas/metabolismo , Ácidos Hidroxâmicos/administração & dosagem , Injeções , Injeções Subcutâneas , Lisina/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Regiões Promotoras Genéticas , Receptores CXCR4/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/metabolismo , Baço/patologia , Regulação para Cima
14.
J Leukoc Biol ; 92(5): 939-50, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22693246

RESUMO

Intraocular tumors reside in an immune-privileged site, yet in certain circumstances, they can undergo immune rejection. Ocular tumor rejection can follow one of two pathways. One pathway is CD4(+) T cell-dependent and culminates in ischemic necrosis of the tumor and phthisis (atrophy) of the eye. A second pathway is also CD4(+) T cell-dependent but does not inflict collateral injury to ocular tissues, and the eye is preserved. We isolated two clones of a murine tumor, Ad5E1 that undergo profoundly different forms of immune rejection in the eye. Clone 2.1 tumors undergo an ischemic necrotizing form of rejection that requires IFN-γ, T cells, and ocular macrophages and culminates in destruction of the eye. By contrast, the second clone of Ad5E1, clone 4, undergoes rejection that also requires T cells and ocular macrophages, but leaves the eye in pristine condition (nonphthisical rejection). Here, we demonstrate that nonphthisical tumor rejection of clone 4 tumors is IFN-γ-independent but requires an ocular macrophage population that contains M1 and M2 macrophages. Clone 4 tumor-bearing eyes displayed ten- and 15-fold increases in M2- and M1-associated markers Arg1 and NO2, respectively. This is in sharp contrast to previous results with clone 2.1 tumor rejection, in which M2 markers were undetectable, and the eye was destroyed. These results suggest that the presence of M2 macrophages tempers the immune rejection of intraocular tumors and promotes immune effectors that inflict minimal injury to innocent bystander cells and thereby preserve the integrity and function of the eye.


Assuntos
Neoplasias Oculares/imunologia , Neoplasias Oculares/patologia , Interferon gama/imunologia , Macrófagos/imunologia , Animais , Modelos Animais de Doenças , Olho/patologia , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
15.
J Leukoc Biol ; 91(3): 475-83, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22180630

RESUMO

Introduction of alloantigens into the AC induces a form of immune tolerance known as ACAID, which induces antigen-specific CD8+ Tregs, contributing to ocular immune privilege by down-regulating immune responses. Recent evidence suggests IFN-γ is needed for the suppressive function of CD8+ ACAID Tregs. This study tested the hypothesis that IFN-γ is needed for alloantigen-specific ACAID CD8+ Tregs to execute their suppressive function but is not required for the establishment of ACAID CD8+ Tregs. To address this hypothesis, ACAID was induced by injecting BALB/c spleen cells into the AC of WT C57BL/6 mice, IFN-γ(-/-) C57BL/6 mice, or anti-IFN-γ-treated WT C57BL/6 mice. LAT assays using C57BL/6 APCs as stimulators, CD4+ T cells from C57BL/6 mice previously immunized toward BALB/c alloantigens as effector cells, and IFN-γ-competent, IFN-γ(-/-), or IFN-γR(-/-) CD8+ Tregs were used to evaluate the suppressive function of CD8+ ACAID Tregs in response to IFN-γ. IFN-γ(-/-) mice or mice treated with anti-IFN-γ antibody prior to AC injection of alloantigen failed to develop ACAID. The suppressive function of IFN-γ(-/-) ACAID CD8+ Tregs was restored through the administration of exogenous IFN-γ. This suppressive responsiveness toward IFN-γ was CD8+ Treg-intrinsic, as CD8+ Tregs from IFN-γR(-/-) mice, which were primed in the AC with alloantigens, were not able to suppress alloantigen-specific DTH responses. These results indicate that IFN-γ is not needed for the induction of CD8+ ACAID Tregs but is required for ACAID Tregs to exert the suppression of allospecific DTH responses.


Assuntos
Câmara Anterior/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/metabolismo , Tolerância Imunológica , Interferon gama/genética , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/metabolismo
16.
Cancer Res ; 71(7): 2445-54, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21307132

RESUMO

Although intraocular tumors reside in an immune privileged site, some tumors are rejected nonetheless. For example, intraocular adenovirus-induced (Ad5E1; adenovirus type 5 early region 1) tumors are rejected in syngeneic C57BL/6 mice by one of two pathways. One pathway leads to extensive necrosis of innocent bystander cells and culminates in destruction of the eye, a condition called phthisis. The second pathway is characterized by piecemeal tumor cell death that rids the eye of the tumor while preserving the architecture and function of the eye. To study the mechanisms of phthisical tumor rejection, we isolated a cell clone-designated clone 2.1 that consistently undergoes rejection in a phthisical manner. CD4(+) T cells and macrophages were required for phthisical rejection of intraocular clone 2.1 tumors and M1 macrophages were involved in mediating tumor rejection. In vitro and in vivo inhibition of iNOS (inducible nitric oxide synthase) abolished macrophage-mediated killing of tumor cells and rejection of intraocular tumors. A role for M1 macrophages was further supported by investigations showing that intraocular tumors grew progressively in IFN-γ KO (knockout) mice. Studies in mice deficient in TNF-α, TNF receptor-1, or TNF receptor-2 revealed that although TNF-α was not needed for tumor rejection, it was required for the development of necrotizing inflammation and phthisis of tumor-bearing eyes. Together, our findings suggest new strategies to successfully eliminate ocular tumors while preserving the integrity of the eye.


Assuntos
Neoplasias Oculares/imunologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adenoviridae/genética , Proteínas E1 de Adenovirus/genética , Animais , Transformação Celular Viral , Inibidores Enzimáticos/farmacologia , Neoplasias Oculares/enzimologia , Neoplasias Oculares/genética , Neoplasias Oculares/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , NG-Nitroarginina Metil Éster/farmacologia , Linfócitos T/imunologia , Transfecção , Fator de Necrose Tumoral alfa/biossíntese
17.
Invest Ophthalmol Vis Sci ; 52(5): 2211-6, 2011 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-21212171

RESUMO

PURPOSE: To determine the function of γδ T cells in early- and late-phase responses in allergic conjunctivitis. METHODS: Wild-type (WT) C57BL/6 and γδ T cell-deficient (TCR-δ(-/-)) mice were immunized intraperitoneally and challenged topically for 7 consecutive days with short ragweed pollen. Natural killer T (NKT) and γδ T cell-double-deficient mice were generated by treating TCR-δ(-/-) mice with anti-CD1d antibody. Allergic conjunctivitis was evaluated clinically, and the late-phase response was assessed by histopathology. Cytokine profiles were evaluated by ELISA. The afferent and efferent arms of allergic conjunctivitis were assessed by adoptive transfer of CD4(+) T cells from WT or TCR-δ(-/-) mice into naive TCR-δ(-/-) or WT mice. RESULTS: TCR-δ(-/-) mice had decreased clinical manifestations of allergic conjunctivitis compared with WT mice. TCR-δ(-/-) mice had decreased eosinophilic infiltration compared with WT mice. TCR-δ(-/-) mice produced less Th2-associated cytokines interleukin (IL)-4, -5, and -13 compared with WT mice. Clinical manifestations of allergic conjunctivitis were lowest in NKT cell-depleted TCR-δ(-/-) mice. However, late-phase allergic conjunctivitis in NKT cell-depleted, TCR-δ(-/-) mice was the same as TCR-δ(-/-) mice. Adoptive transfer of CD4(+) T cells revealed that γδ T cells are needed for the afferent and efferent arms of allergic conjunctivitis. CONCLUSIONS: γδ T cells are needed for full expression of both the clinical manifestations and the late phase of allergic conjunctivitis. Thus, γδ T cells have an important impact in the expression of allergic conjunctivitis and are a potential therapeutic target in the management of allergic diseases of the ocular surface.


Assuntos
Conjuntivite Alérgica/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/fisiologia , Linfócitos T/imunologia , Transferência Adotiva , Ambrosia , Animais , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/imunologia , Pólen , Células Th2/imunologia
18.
Invest Ophthalmol Vis Sci ; 52(6): 3094-102, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21330669

RESUMO

PURPOSE: To explore the role of natural killer T (NKT) cells in the development of liver metastases in mice harboring intraocular melanomas. METHODS: Cells derived from the cutaneous B16 melanoma cell line (B16LS9) were transplanted either into the vitreous body or under the spleen capsules of wild-type C57BL/6 mice and NKT-cell-deficient Jα18(-/-) and CD1d(-/-) mice. The development of liver metastases was evaluated by histopathology. The effect of NK cells on liver metastases was determined by selective depletion with anti-asialo-GM1 antiserum in vivo and NK-cell-mediated cytolysis of B16LS9 melanoma cells in vitro. The role of IL-10 and transforming growth factor (TGF)-ß in the inhibition of liver NK resistance to liver metastases was determined by in vivo and in vitro neutralization with monoclonal antibodies. RESULTS: Liver NKT cells, especially type I NKT cells, enhanced liver metastases arising from intraocular melanomas. NKT-cell-deficient mice developed significantly fewer liver metastases that were NK-cell dependent. Tumor-induced liver NKT cells, especially type I NKT cells, inhibited liver NK-cell cytotoxicity by an IL-10-dependent process. CONCLUSIONS: NKT cells exert protective effects in many murine tumor models. However, the present results reveal that NKT cells exacerbate liver metastases arising from intraocular melanomas. To the authors' knowledge, this is the first report that liver NKT cells, especially type I NKT cells, inhibit liver NK-cell antimetastatic activity by the production of IL-10. These results suggest that hepatic NKT cell activity can have an important effect in the immune surveillance of liver metastases.


Assuntos
Neoplasias Oculares/patologia , Neoplasias Hepáticas/secundário , Melanoma Experimental/secundário , Células T Matadoras Naturais/imunologia , Neoplasias Esplênicas/patologia , Corpo Vítreo/patologia , Animais , Citotoxicidade Imunológica , Ensaio de Imunoadsorção Enzimática , Neoplasias Oculares/imunologia , Feminino , Citometria de Fluxo , Gangliosídeo G(M1)/imunologia , Interleucina-10/fisiologia , Fígado/imunologia , Neoplasias Hepáticas/imunologia , Masculino , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esplenectomia , Neoplasias Esplênicas/imunologia , Fator de Crescimento Transformador beta/fisiologia , Corpo Vítreo/imunologia
19.
Ocul Immunol Inflamm ; 18(2): 80-90, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20370332

RESUMO

Mechanisms that maintain ocular immune privilege may contribute to ocular tumor progression by inhibiting tumoricidal immune responses. Consistent with that notion are observations from transplantable tumor models in mice demonstrating that the tumoricidal activity of CD8(+) cytolytic T lymphocytes (CTL) may be inhibited directly by interfering with CTL effector function in the eye or indirectly by abrogating the effector function of CD8+ T cell-activated intratumoral macrophages that are critical for ocular tumor rejection. In addition, epigenetic gene regulation by factors within the ocular tumor environment favors the generation of tumor variants that are resistant to CD8(+) CTL. Intratumoral macrophages may be essential for eliminating these variants because, unlike CTL, their tumoricidal activity is nonspecific. Hence, the inhibition of macrophage effector function within the eye, presumably to preserve immune privilege by minimizing ocular immunopathology, may hasten the outgrowth of tumor escape variants which contributes to ocular tumor progression.


Assuntos
Neoplasias Oculares/imunologia , Melanoma/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Epigênese Genética , Olho/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Tolerância Imunológica , Macrófagos/imunologia , Camundongos , Linfócitos T Citotóxicos/imunologia , Evasão Tumoral/imunologia
20.
Discov Med ; 10(54): 452-61, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21122477

RESUMO

Corneal transplantation is the oldest, most common, and arguably the most successful form of organ transplantation. In uncomplicated first-time cases, corneal allografts enjoy a success rate of up to 90% even though the transplants are performed without HLA matching or the use of systemic immunosuppressive drugs. In rodents, corneal allografts transplanted across entire MHC and multiple minor histocompatibility barriers enjoy long-term survival in >50% of the hosts, while skin grafts invariably undergo immune rejection. These observations are the basis for "immune privilege" of corneal transplants. In spite of this immune privilege, immune rejection can occur and remains the leading cause of corneal graft failure. Rodent models of penetrating keratoplasty have facilitated studies that have challenged, and in some cases, refuted prevailing dogmas. The long-held belief that CD4+ T helper 1 (Th1) cells were the sole mediators of corneal allograft rejection has fallen to the wayside based on studies in interferon-gamma (IFN-γ)⁻(/)⁻ mice. The dogma that biasing the alloimmune response down a Th2 pathway would enhance graft survival has also been disproven, and in fact, compelling evidence indicates that Th2-based immune rejection of corneal allografts is swifter and more intense than Th1-based rejection. Animal studies have also pre-empted emerging dogmas including the hypothesis that Th17 cells play a crucial role in allograft rejection. Instead, IL-17A appears to be necessary for corneal allograft survival. Finally, IFN-γ, and IL-17A, which were normally viewed as proinflammatory, exert the opposite effect in the context of corneal transplantation and are necessary for corneal allograft survival.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Transplante de Córnea , Animais , Antígenos CD/imunologia , Rejeição de Enxerto/imunologia , Humanos , Imunidade Celular , Interleucina-17/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA