Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
bioRxiv ; 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38168302

RESUMO

Background: The inability to evaluate host immunity in a rapid quantitative manner in patients with sepsis has severely hampered development of novel immune therapies. The ELISpot assay is a functional bioassay that measures the number of cytokine-secreting cells and the relative amount of cytokine produced at the single-cell level. A key advantage of ELISpot is its excellent dynamic range enabling a more precise quantifiable assessment of host immunity. Herein, we tested the hypothesis on whether the ELISpot assay can detect dynamic changes in both innate and adaptive immunity as they often occur during sepsis. We also tested whether ELISpot could detect the effect of immune drug therapies to modulate innate and adaptive immunity. Methods: Mice were made septic using sublethal cecal ligation and puncture (CLP). Blood and spleens were harvested serially and ex vivo IFN-γ and TNF-α production were compared by ELISpot and ELISA. The capability of ELISpot to detect changes in innate and adaptive immunity due to in vivo immune therapy with dexamethasone, IL-7, and arginine was also evaluated. Results: ELISpot confirmed a decreased innate and adaptive immunity responsiveness during sepsis progression. More importantly, ELISpot was also able to detect changes in adaptive and innate immunity in response to immune-modulatory reagents, for example dexamethasone, arginine, and IL-7 in a readily quantifiable manner, as predicted by the reagents known mechanisms of action. ELISpot and ELISA results tended to parallel one another although some differences were noted. Conclusion: ELISpot offers a unique capability to assess the functional status of both adaptive and innate immunity over time. The results presented herein demonstrate that ELISpot can also be used to detect and follow the in vivo effects of drugs to ameliorate sepsis-induced immune dysfunction. This capability would be a major advance in guiding new immune therapies in sepsis.

2.
Nat Med ; 5(2): 157-63, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9930862

RESUMO

To evaluate the utility of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) as a cancer therapeutic, we created leucine zipper (LZ) forms of human (hu) and murine (mu) TRAIL to promote and stabilize the formation of trimers. Both were biologically active, inducing apoptosis of both human and murine target cells in vitro with similar specific activities. In contrast to the fulminant hepatotoxicity of LZ-huCD95L in vivo, administration of either LZ-huTRAIL or LZ-muTRAIL did not seem toxic to normal tissues of mice. Finally, repeated treatments with LZ-huTRAIL actively suppressed growth of the TRAIL-sensitive human mammary adenocarcinoma cell line MDA-231 in CB.17 (SCID) mice, and histologic examination of tumors from SCID mice treated with LZ-huTRAIL demonstrated clear areas of apoptotic necrosis within 9-12 hours of injection.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Relação Dose-Resposta a Droga , Proteína Ligante Fas , Humanos , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/síntese química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Conformação Proteica , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/síntese química
3.
J Exp Med ; 179(5): 1597-604, 1994 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8163939

RESUMO

Contact hypersensitivity (CHS) responses require the participation of T cells, along with a variety of cytokines and adhesion molecules. In the classical CHS, antigen-specific T cells are recruited to a site of antigenic challenge, where they react with antigen, release cytokines, and attract other inflammatory cells. In the mouse model of CHS, this reaction is elicited in sensitized mice by application of the immunogen 4-7 d after immunization. The reaction peaks at 24 h, is slightly reduced by 48 h, and can return to normal by 72 h. This is in spite of the fact that some antigen is still present at the site of challenge. Here we examined the hypothesis that locally produced interleukin 10 (IL-10) regulates the duration of the response. Our data show that IL-10 protein peaked 10-14 h after antigenic challenge and returned to background by 24 h. The production of IL-10 protein corresponded with, and followed IL-10 mRNA transcription as detected by reverse transcriptase-polymerase chain reaction. During peak IL-10 production after antigenic challenge, it was not possible to transfer CHS with immune lymphoid cells, unless neutralizing antibody to IL-10 was given first. Additionally, when sensitized mice were given neutralizing anti-IL-10 antibody at the time of antigenic challenge, the duration of CHS was prolonged well beyond the natural course of the response. Finally, we demonstrate that rIL-10, when injected into the skin before antigenic challenge, prevented the elicitation of CHS in previously sensitized mice. Taken together, our data show an important role for IL-10 in the natural regulation of CHS responses in vivo.


Assuntos
Dermatite de Contato/imunologia , Interleucina-10/imunologia , Linfócitos T/imunologia , Animais , Sequência de Bases , DNA , Interleucina-10/análise , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Pele/imunologia
4.
J Exp Med ; 190(8): 1155-64, 1999 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-10523613

RESUMO

TRAIL (TNF-related apoptosis-inducing ligand) is a member of the TNF family that induces apoptosis in a variety of cancer cells. In this study, we demonstrate that human CD11c(+) blood dendritic cells (DCs) express TRAIL after stimulation with either interferon (IFN)-gamma or -alpha and acquire the ability to kill TRAIL-sensitive tumor cell targets but not TRAIL-resistant tumor cells or normal cell types. The DC-mediated apoptosis was TRAIL specific, as soluble TRAIL receptor blocked target cell death. Moreover, IFN-stimulated interleukin (IL)-3 receptor (R)alpha(+) blood precursor (pre-)DCs displayed minimal cytotoxicity toward the same target cells, demonstrating a clear functional difference between the CD11c(+) DC and IL-3Ralpha(+) pre-DC subsets. These results indicate that TRAIL may serve as an innate effector molecule on CD11c(+) DCs for the elimination of spontaneously arising tumor cells and suggest a means by which TRAIL-expressing DCs may regulate or eliminate T cells responding to antigen presented by the DCs.


Assuntos
Apoptose/efeitos dos fármacos , Células Dendríticas/metabolismo , Glicoproteínas de Membrana/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apresentação de Antígeno/imunologia , Antígenos CD/imunologia , Proteínas Reguladoras de Apoptose , Citocinas/farmacologia , Citotoxicidade Imunológica , Citometria de Fluxo , Histocitoquímica , Humanos , Integrina alfaXbeta2/imunologia , Interferons/farmacologia , Receptores de Interleucina-3/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF
5.
J Exp Med ; 188(5): 887-96, 1998 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-9730890

RESUMO

Apoptosis is critical to homeostasis of multicellular organisms. In immune privileged sites such as the eye, CD95 ligand (FasL)-induced apoptosis controls dangerous inflammatory reactions that can cause blindness. Recently, we demonstrated that apoptotic cell death of inflammatory cells was a prerequisite for the induction of immune deviation after antigen presentation in the eye. In this report, we examine the mechanism by which this takes place. Our results show that Fas- mediated apoptosis of lymphoid cells leads to rapid production of interleukin (IL)-10 in these cells. The apoptotic cells containing IL-10 are responsible for the activation of immune deviation through interaction with antigen-presenting cells (APC). In support of this, we found that apoptotic cells from IL-10(+/+) animals fed to APC in vitro promote Th2 cell differentiation, whereas apoptotic IL-10(-/-) cells, as well as nonapoptotic cells, favor Th1 induction. Thus, apoptotic cell death and tolerance are linked through the production of an antiinflammatory cytokine to prevent dangerous and unwanted immune responses that might compromise organ integrity.


Assuntos
Apoptose/imunologia , Glicoproteínas de Membrana/fisiologia , Receptor fas/fisiologia , Animais , Câmara Anterior/imunologia , Câmara Anterior/metabolismo , Apresentação de Antígeno , Diferenciação Celular/imunologia , Proteína Ligante Fas , Inflamação/imunologia , Inflamação/prevenção & controle , Interleucina-10/biossíntese , Interleucina-10/genética , Interleucina-4/genética , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Th2/citologia , Células Th2/metabolismo
6.
J Exp Med ; 189(8): 1343-54, 1999 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-10209050

RESUMO

TRAIL (tumor necrosis factor [TNF]-related apoptosis-inducing ligand) is a molecule that displays potent antitumor activity against selected targets. The results presented here demonstrate that human monocytes rapidly express TRAIL, but not Fas ligand or TNF, after activation with interferon (IFN)-gamma or -alpha and acquire the ability to kill tumor cells. Monocyte-mediated tumor cell apoptosis was TRAIL specific, as it could be inhibited with soluble TRAIL receptor. Moreover, IFN stimulation caused a concomitant loss of TRAIL receptor 2 expression, which coincides with monocyte acquisition of resistance to TRAIL-mediated apoptosis. These results define a novel mechanism of monocyte-induced cell cytotoxicity that requires TRAIL, and suggest that TRAIL is a key effector molecule in antitumor activity in vivo.


Assuntos
Antineoplásicos/metabolismo , Glicoproteínas de Membrana/metabolismo , Monócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/farmacologia , Fosfatidilserinas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
7.
Science ; 270(5239): 1189-92, 1995 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-7502042

RESUMO

The eye is a privileged site that cannot tolerate destructive inflammatory responses. Inflammatory cells entering the anterior chamber of the eye in response to viral infection underwent apoptosis that was dependent on Fas (CD95)-Fas ligand (FasL) and produced no tissue damage. In contrast, viral infection in gld mice, which lack functional FasL, resulted in an inflammation and invasion of ocular tissue without apoptosis. Fas-positive but not Fas-negative tumor cells were killed by apoptosis when placed within isolated anterior segments of the eyes of normal but not FasL-negative mice. FasL messenger RNA and protein were detectable in the eye. Thus, Fas-FasL interactions appear to be an important mechanism for the maintenance of immune privilege.


Assuntos
Câmara Anterior/imunologia , Apoptose , Tolerância Imunológica , Glicoproteínas de Membrana/fisiologia , Animais , Câmara Anterior/virologia , Sequência de Bases , Olho/metabolismo , Proteína Ligante Fas , Expressão Gênica , Ceratite Herpética/imunologia , Leucemia L1210 , Linfócitos/citologia , Linfócitos/imunologia , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neutrófilos/citologia , Neutrófilos/imunologia , RNA Mensageiro/análise , RNA Mensageiro/genética , Células Tumorais Cultivadas , Receptor fas/fisiologia
8.
J Clin Invest ; 99(3): 396-402, 1997 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9022072

RESUMO

Although anatomical barriers and soluble mediators have been implicated in immune privilege, it appears that the apoptotic cell death of Fas+ cells by tissue-associated CD95 ligand (Fas ligand, FasL) is an important component. One clinical example of the function of an immune privileged site is the success of human corneal transplants, where a very high percentage of transplants accept without tissue matching or immunosuppressive therapy. Since the mouse cornea expresses abundant Fas ligand and immune privilege has been implicated in the success of these transplants, we examined the role of FasL in corneal transplantation. Our results show that human corneas express functional FasL capable of killing Fas+ lymphoid cells in an in vitro culture system. Using a mouse model for corneal allograft transplantation, FasL+ orthografts were accepted at a rate of 45%, whereas FasL- grafts, or normal grafts transplanted to Fas- mice, were rejected 100% of the time. Histological analysis found that FasL+ grafts contained apoptotic mononuclear cells indicating the induction of apoptosis by the graft, while rejecting FasL- corneas contained numerous inflammatory cells without associated apoptosis. Taken together our results demonstrate that FasL expression on the cornea is a major factor in corneal allograft survival and, thus, we provide an explanation for one of the most successful tissue transplants performed in humans.


Assuntos
Transplante de Córnea/imunologia , Sobrevivência de Enxerto/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/fisiologia , Transplante Homólogo/imunologia , Receptor fas/imunologia , Actinas/genética , Adolescente , Adulto , Idoso , Animais , Apoptose/imunologia , Criança , Pré-Escolar , Córnea/imunologia , Citotoxicidade Imunológica , Proteína Ligante Fas , Feminino , Rejeição de Enxerto/imunologia , Células HeLa , Teste de Histocompatibilidade , Humanos , Imuno-Histoquímica , Terapia de Imunossupressão , Lactente , Recém-Nascido , Inflamação , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Mensageiro/análise
9.
Curr Opin Immunol ; 10(5): 559-63, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9794836

RESUMO

Apoptosis research is benefiting from bioinformatic approaches to identify new components of the cell death machinery and novel cell death inducers/receptors. Over the past year, knowledge of the system involving TNF-related apoptosis-inducing ligand (TRAIL) and its receptors has increased via genomic database analysis to include four distinct receptors that interact with a single ligand. Currently, these molecules are of major interest due to their potential roles and application in cancer therapy.


Assuntos
Apoptose , Glicoproteínas de Membrana/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Proteínas Ligadas por GPI , Humanos , RNA Mensageiro/análise , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/genética , Membro 10c de Receptores do Fator de Necrose Tumoral , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Receptores Chamariz do Fator de Necrose Tumoral
10.
Cancer Gene Ther ; 13(6): 628-32, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16456549

RESUMO

Renal cell carcinoma (RCC) will cause greater than 12,000 deaths in the United States this year. The lack of effective therapy for disseminated RCC has stimulated the search for novel treatments including immunotherapeutic strategies, but poor therapeutic responses and marked toxicity have limited their use. The tumor necrosis factor (TNF) family member TNF-related apoptosis-inducing ligand (TRAIL)/Apo-2L induces apoptosis in various tumor cell types, while having little cytotoxicity against normal cells. In this study, we investigated the tumoricidal potential of a recombinant adenovirus encoding human TNFSF10 (Ad5-TRAIL), alone and in combination with a panel of histone deacetylase inhibitors (HDACi), against the TRAIL/Apo-2L-resistant RCC line 786-O and normal human renal proximal tubule epithelial cells (RPTEC). Ad5-TRAIL was unable to induce apoptosis in either 786-O or RPTEC alone; however, tumor cell apoptosis occurred when Ad5-TRAIL was combined with HDAC inhibition. Except when combined with trichostatin A, RPTEC were not sensitized to Ad5-TRAIL by HDACi. In 786-O, HDAC inhibition induced CAR expression, permitting increased adenoviral infection and transgene expression. It also induced TRAIL-R2 expression, accelerated the death-inducing signaling complex formation and enhanced caspase-8 activation. Our results demonstrate the utility of combining Ad5-TRAIL with HDACi against RCC, and mechanistically define how this combination modulates RCC sensitivity to TRAIL/Apo-2L and adenoviral infection.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Inibidores de Histona Desacetilases , Neoplasias Renais/tratamento farmacológico , Glicoproteínas de Membrana/genética , Fator de Necrose Tumoral alfa/genética , Adenoviridae/classificação , Adenoviridae/genética , Antineoplásicos/administração & dosagem , Proteínas Reguladoras de Apoptose/administração & dosagem , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Caspase 8 , Caspases/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Inibidores Enzimáticos/uso terapêutico , Vetores Genéticos/genética , Histona Desacetilases/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Glicoproteínas de Membrana/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/administração & dosagem
11.
J Natl Cancer Inst ; 93(13): 998-1007, 2001 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-11438565

RESUMO

BACKGROUND: Immunization with modified tumor cells carrying recombinant immunomodulatory genes is being explored as cancer immunotherapy. In this study, we examine whether canarypox ALVAC viruses carrying immunostimulatory cytokine genes (granulocyte-macrophage colony-stimulating factor, interleukin 2, interleukin 12, and tumor necrosis factor-alpha) can induce antitumor immunity (to rechallenge) in the RM-1 model of a highly aggressive, weakly immunogenic murine prostate cancer. METHODS: For antitumor activity studies, RM-1 murine prostate cancer cells were infected with the parental ALVAC virus or one or two recombinant ALVAC-cytokine viruses and then injected into male C57BL/6 mice. For rechallenge studies, other mice were first given an injection subcutaneously with irradiated (nonproliferating) recombinant ALVAC-infected RM-1 cells and then (10 days later) with untreated RM-1 cells. For the determination of which immune cells were required for antitumor activity, mice were immunodepleted of CD4, CD8, or natural killer (NK) NK1.1 cells with the corresponding monoclonal antibodies and were then given an injection of ALVAC-cytokine-infected RM-1 cells. For all experiments, tumor outgrowth and animal survival were monitored. RESULTS: After subcutaneous injection into mice, RM-1 cells infected with one (except ALVAC-interleukin 2) or two ALVAC-cytokine recombinants had statistically significantly greater antitumor activity than RM-1 cells infected with parental ALVAC (P<.001 for all; two-sided test). The antitumor activity of RM-1 cells infected with any two ALVAC-cytokine recombinants was greater than, but not statistically significantly different from, that of RM-1 cells infected with any one ALVAC-cytokine recombinant. NK1.1 cells were necessary for antitumor activity, but tumor-specific CD4(+) regulatory T cells were also induced that inhibited CD8(+) RM-1-specific cytotoxic T cells, resulting in the lack of immunity to a rechallenge by RM-1 cells. DISCUSSION: Canarypox viruses can transfer immunostimulatory cytokine genes into RM-1 prostate cancer cells. When such cells were injected into mice, the cytokines induced an antitumor response against this highly aggressive, weakly immunogenic tumor. This response, however, did not protect the mouse against a rechallenge with RM-1 cells because suppressor CD4(+) T cells were induced that inhibited tumor-specific CD8(+) cytotoxic T cells.


Assuntos
Avipoxvirus/genética , Neoplasias da Próstata/terapia , Proteínas , Animais , Anticorpos Monoclonais/metabolismo , Antígenos/biossíntese , Antígenos Ly , Antígenos de Superfície , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Citometria de Fluxo , Técnicas de Transferência de Genes , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interleucina-12/genética , Interleucina-2/genética , Lectinas Tipo C , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Transplante de Neoplasias , Neoplasias da Próstata/imunologia , Biossíntese de Proteínas , Proteínas Recombinantes/metabolismo , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/genética
13.
Cancer Gene Ther ; 14(12): 976-84, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17853923

RESUMO

High levels of decoy receptor 2 (DcR2; TRAIL-R4) expression are correlated with TRAIL resistance in prostate cancer cells. In addition, upregulation of TRAIL death receptor (DR4 and DR5) expression, either by ionizing radiation or chemotherapy, can sensitize cancer cells to TRAIL. Considering more than half of human cancers are TRAIL resistant, modulation of surface TRAIL receptor expression appears to be an attractive treatment modality to counteract TRAIL resistance. In this study, three siRNA duplexes targeting DcR2 receptor were tested. Ad5hTRAIL infections were performed to overexpress human full-length TRAIL to induce cell death, and the in vitro tumorigenic potential of prostate cancer cells was assessed using colony-forming assays on soft agar. The DU145 and LNCaP prostate cancer cell lines, which express high levels of DcR2, were resistant to Ad5hTRAIL-induced death. Downregulation of surface DcR2 expression by siRNA sensitized these prostate cancer cell lines to Ad5hTRAIL. In addition, DcR2 siRNA-mediated knockdown of DcR2, followed by Ad5hTRAIL infection, dramatically reduced the in vitro tumorigenic potential of prostate cancer cells. Collectively, our results suggest the potential for combining receptor-specific siRNA with TRAIL in the treatment of certain cancers.


Assuntos
Adenoviridae , Terapia Genética , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias da Próstata/metabolismo , RNA Interferente Pequeno/biossíntese , Transdução Genética , Receptores Chamariz do Fator de Necrose Tumoral/antagonistas & inibidores , Animais , Células COS , Morte Celular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral/biossíntese , Receptores Chamariz do Fator de Necrose Tumoral/genética , Regulação para Cima/genética
14.
Immunol Rev ; 156: 167-84, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9176707

RESUMO

Immune privilege is a term applied to several organs that have a unique relationship with the immune response. These sites prohibit the spread of inflammation since even minor episodes can threaten organ integrity and function. The most prominent examples of these are the eye, brain and reproductive organs where immune responses either do not proceed, or proceed in a manner different from other areas. Once thought to be a passive process relying on physical barriers, immune privilege can now be viewed as an active process that utilizes multiple mechanisms to maintain organ function. Recently there has been a renewed interest in immune privilege when it was shown that two privileged sites (the eye and testes) constitutively express FasL, which functions by killing lymphoid cells that invade these areas. Here we will examine the role of FasL in immune privilege and discuss how this molecule interacts with other elements of the inflammatory response to maintain organ integrity in the face of potentially damaging immune reactions.


Assuntos
Morte Celular , Olho/imunologia , Glicoproteínas de Membrana/imunologia , Receptor fas/imunologia , Animais , Adesão Celular , Olho/citologia , Olho/ultraestrutura , Proteína Ligante Fas , Previsões , Humanos , Tecido Linfoide , Neuropeptídeos/imunologia , Substância P/imunologia , Imunologia de Transplantes , Peptídeo Intestinal Vasoativo/imunologia
15.
Mol Ther ; 4(3): 257-66, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11545617

RESUMO

TRAIL is a member of the tumor necrosis factor superfamily that induces apoptosis in a variety of tumor cell types both in vitro and in vivo, while demonstrating minimal cytotoxicity toward normal tissues. One disadvantage to previous in vivo protocols was the need for large quantities of TRAIL to suppress tumor growth. Here we engineered a replication-deficient adenovirus to encode human TNFSF10 (Ad5-TRAIL) as an alternative to recombinant, soluble TRAIL protein. The results show that TRAIL-sensitive prostate tumor cell targets infected with Ad5-TRAIL undergo apoptosis through the production and expression of TRAIL protein. This activity was limited to TRAIL-sensitive tumor cells, as normal prostate epithelial cells were not killed by Ad5-TRAIL. Furthermore, in vivo administration of Ad5-TRAIL at the site of tumor implantation suppressed the outgrowth of human prostate tumor xenografts in SCID mice. Histologic examination of prostate tumors treated locally with Ad5-TRAIL revealed areas of apoptosis within 24 hours of injection. These results further define Ad5-TRAIL as a novel anti-tumor therapeutic and demonstrate its potential use as a means for treating prostate tumors, as well as other solid tumors, in vivo.


Assuntos
Adenoviridae/genética , Apoptose , Terapia Genética/métodos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/uso terapêutico , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Proteínas Reguladoras de Apoptose , Vetores Genéticos/genética , Humanos , Injeções Intralesionais , Masculino , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/metabolismo , Camundongos , Neoplasias da Próstata/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Tempo , Transgenes/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo
16.
Int Immunol ; 7(10): 1617-25, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8562507

RESUMO

Injection of antigen into the anterior chamber (AC) of the eye results in the induction of immune deviation in which antibody production is activated and delayed-type hypersensitivity (DTH) is inhibited. This system is termed anterior chamber associated immune deviation (ACAID) and the model is used to examine certain aspects of the immunologic privilege of the eye. Recent studies have established that following antigen presentation in the eye, an 'ACAID-inducing' signal is produced that directly enters the blood. This signal then homes to the spleen where T cells that down-regulate DTH are activated. For many antigens this 'ACAID signal' is a soluble protein released within 2 days of AC injection. Although the presence of this molecule (or molecules) has been described using several antigens, the exact nature of the soluble mediator has escaped characterization. We have further explored the nature of this signal using HSV-1-induced immune deviation. Our results show the soluble 'signal' was released by T cells that encounter antigen in the ocular microenvironment. This mediator was antigen specific, contained TCR alpha-chain (but not the TCR beta-chain) determinants and had an apparent molecular weight of 46 kDa. These results show that the release of soluble TCR alpha-chain from sites of T cell interaction within the microenvironment of the eye can regulate systemic immune responses. These results have implications for the control of immune response that might be damaging to organs such as the eye.


Assuntos
Apresentação de Antígeno/fisiologia , Fatores Biológicos/imunologia , Olho/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/química , Animais , Câmara Anterior/imunologia , Formação de Anticorpos , Antígenos/administração & dosagem , Antígenos/imunologia , Antígenos Virais/imunologia , Fatores Biológicos/sangue , Fatores Biológicos/química , Barreira Hematorretiniana , Chlorocebus aethiops , Cricetinae , Hipersensibilidade Tardia/imunologia , Tolerância Imunológica , Imunidade Celular , Injeções , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Simplexvirus/imunologia , Solubilidade , Baço/citologia , Baço/imunologia , Esplenectomia , Subpopulações de Linfócitos T/imunologia , Células Vero
17.
J Immunol ; 155(4): 1746-56, 1995 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-7543531

RESUMO

Ag presentation via the anterior chamber of the eye results in a form of immune deviation termed anterior chamber-associated immune deviation (ACAID). The hallmarks of this response are the suppression of delayed-type hypersensitivity with simultaneous induction of Ab production. In this study, we examined the role of the neuropeptides vasoactive intestinal peptide (VIP) and substance P (SP) and found that the levels of these two peptides are controlled by neurogenic stimulation of the eye by light, and that these molecules determine the outcome of Ag presentation in the eye. Mice reared under diurnal conditions had VIP in the iris and ciliary body (not free in aqueous humor) and low levels of SP. Mice that were reared in the dark (or dark-adapted) did not contain detectable levels of VIP but had high levels of SP. The adaptation of diurnal mice to the dark eliminated VIP and increased SP, while adapting dark-reared mice to the diurnal cycle increased VIP and reduced SP. We then tested the hypothesis that immune reactions resulting from Ag presentation in the eye were linked to SP and VIP. We found that a VIP receptor antagonist, when injected into the eye with Ag, reversed ACAID in diurnal mice, while a SP receptor antagonist restored ACAID to dark-adapted mice. We further determined that injection of Ag or TNF-alpha induced VIP release, while SP was liberated into the aqueous humor following reexposure of dark-reared mice to light. Our results demonstrate a close linkage of intraocular immune reactions to neuropeptide levels in the eye.


Assuntos
Câmara Anterior/imunologia , Substância P/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia , Animais , Tolerância Imunológica , Interleucina-2/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Substância P/análise , Linfócitos T/imunologia , Peptídeo Intestinal Vasoativo/análise
18.
Immunity ; 5(1): 7-16, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8758890

RESUMO

We examined the relationship between cell death and tolerance induction following antigen injection into the anterior chamber of the eye. Our data show that when inflammatory cells undergo apoptosis following infection with HSV-1, tolerance to the virus was observed. In contrast, when cell death was absent due to defects in Fas or FasL, immune tolerance was not observed. Further studies revealed that cell death and tolerance required that the lymphoid cells be Fas+ and the eye be FasL+. Additionally, we show that while Fas/FasL-mediated apoptosis occurred in the eye, it was apoptotic cell death that was critical for tolerance induction. Our results further demonstrate immune privilege is not a passive process involving physical barriers, but is an active process that employs an important natural mechanism to induce cell death and immune tolerance.


Assuntos
Apoptose/imunologia , Olho/imunologia , Tolerância Imunológica , Linfócitos/imunologia , Receptor fas/farmacologia , Animais , Apoptose/genética , Transplante de Medula Óssea/imunologia , Proteína Ligante Fas , Herpesvirus Humano 1/imunologia , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/genética , Ligantes , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Quimera por Radiação/imunologia , Receptor fas/genética
19.
Behring Inst Mitt ; (97): 161-74, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8950474

RESUMO

Peripheral deletion of activated T cells has an important function in the regulation of the extent of an immune response. Upon restimulation through the T cell receptor previously stimulated cells have been shown to die by activation-induced cell death. Recent data indicate that this process is mediated by a CD95 (Fas/APO-1)/CD95 ligand interaction which induces apoptosis of the T cell. CD95 ligand (CD95-L) is absent on unactivated T cells but is readily expressed upon stimulation. Here we discuss evidence that CD95-L expression is induced by T cell receptor-mediated signals and is regulated at different levels. Different inhibitors of activation-induced cell death have been found to directly or indirectly act on the signal transduction pathway leading to CD95-L expression. CD95-L seems not only to be induced in T cells after activation but is also found constitutively expressed in many non-lymphoid tissues. This indicates that CD95-L is not only critically involved in activation-induced T cell death, but may have other functions as well. One such function is in the maintenance of immunological privilege, the protection of some tissues from potentially destructive immune responses. Thus, the regulation of CD95 expression in lymphoid and non-lymphoid cells appears to represent a key element in immune regulation.


Assuntos
Apoptose , Regulação da Expressão Gênica , Glicoproteínas de Membrana/biossíntese , Receptor fas/fisiologia , Animais , Calcineurina , Proteínas de Ligação a Calmodulina/fisiologia , Proteína Ligante Fas , Humanos , Modelos Imunológicos , Fosfoproteínas Fosfatases/fisiologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais , Linfócitos T/imunologia
20.
J Immunol ; 162(9): 5205-11, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10227994

RESUMO

Activation-induced cell death of peripheral T cells results from the interaction between Fas and Fas ligand. Resting peripheral T cells are resistant to Fas-induced apoptosis and become susceptible only after their activation. We have investigated the molecular mechanism mediating the sensitization of resting peripheral T cells to Fas-mediated apoptosis following TCR stimulation. TCR activation decreases the steady state protein levels of FLIP (FLICE-like inhibitory protein), an inhibitor of the Fas signaling pathway. Reconstitution of intracellular FLIP levels by the addition of a soluble HIV transactivator protein-FLIP chimera completely restores resistance to Fas-mediated apoptosis in TCR primary T cells. Inhibition of IL-2 production by cyclosporin A, or inhibition of IL-2 signaling by rapamycin or anti-IL-2 neutralizing Abs prevents the decrease in FLIP levels and confers resistance to Fas-mediated apoptosis following T cell activation. Using cell cycle-blocking agents, we demonstrate that activated T cells arrested in G1 phase contain high levels of FLIP protein, whereas activated T cells arrested in S phase have decreased FLIP protein levels. These findings link regulation of FLIP protein levels with cell cycle progression and provide an explanation for the increase in TCR-induced apoptosis observed during the S phase of the cell cycle.


Assuntos
Apoptose/imunologia , Proteínas de Transporte/metabolismo , Ciclo Celular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Receptor fas/fisiologia , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/antagonistas & inibidores , Células Cultivadas , Ciclosporina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Humanos , Soros Imunes/farmacologia , Imunidade Inata , Interleucina-2/antagonistas & inibidores , Interleucina-2/biossíntese , Interleucina-2/imunologia , Ativação Linfocitária/efeitos dos fármacos , Dados de Sequência Molecular , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sirolimo/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa