Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nat Med ; 6(4): 443-6, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742152

RESUMO

Inhibitory receptors have been proposed to modulate the in vivo cytotoxic response against tumor targets for both spontaneous and antibody-dependent pathways. Using a variety of syngenic and xenograft models, we demonstrate here that the inhibitory FcgammaRIIB molecule is a potent regulator of antibody-dependent cell-mediated cytotoxicity in vivo, modulating the activity of FcgammaRIII on effector cells. Although many mechanisms have been proposed to account for the anti-tumor activities of therapeutic antibodies, including extended half-life, blockade of signaling pathways, activation of apoptosis and effector-cell-mediated cytotoxicity, we show here that engagement of Fcgamma receptors on effector cells is a dominant component of the in vivo activity of antibodies against tumors. Mouse monoclonal antibodies, as well as the humanized, clinically effective therapeutic agents trastuzumab (Herceptin(R)) and rituximab (Rituxan(R)), engaged both activation (FcgammaRIII) and inhibitory (FcgammaRIIB) antibody receptors on myeloid cells, thus modulating their cytotoxic potential. Mice deficient in FcgammaRIIB showed much more antibody-dependent cell-mediated cytotoxicity; in contrast, mice deficient in activating Fc receptors as well as antibodies engineered to disrupt Fc binding to those receptors were unable to arrest tumor growth in vivo. These results demonstrate that Fc-receptor-dependent mechanisms contribute substantially to the action of cytotoxic antibodies against tumors and indicate that an optimal antibody against tumors would bind preferentially to activation Fc receptors and minimally to the inhibitory partner FcgammaRIIB.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD/imunologia , Linfoma de Células B/imunologia , Melanoma Experimental/imunologia , Receptor ErbB-2/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Anticorpos Monoclonais Murinos , Antígenos CD/genética , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Feminino , Humanos , Imunização Passiva , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores de IgG/genética , Rituximab , Trastuzumab
2.
J Exp Med ; 185(11): 1977-85, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9166427

RESUMO

Interleukin (IL)-12 is a monocyte- and macrophage-derived cytokine that plays a crucial role in both the innate and the acquired immune response. In this study, we examined the effects that ligating specific macrophage receptors had on the induction of IL-12 by lipopolysaccharide (LPS). We report that ligation of the macrophage Fcgamma, complement, or scavenger receptors inhibited the induction of IL-12 by LPS. Both mRNA synthesis and protein secretion were diminished to near-undetectable levels following receptor ligation. Suppression was specific to IL-12 since IL-10 and tumor necrosis factor-alpha (TNF-alpha) production were not inhibited by ligating macrophage receptors. The results of several different experimental approaches suggest that IL-12 downregulation was due to extracellular calcium influxes that resulted from receptor ligation. First, preventing extracellular calcium influxes, by performing the assays in EGTA, abrogated FcgammaR-mediated IL-12(p40) mRNA suppression. Second, exposure of macrophages to the calcium ionophores, ionomycin or A23187, mimicked receptor ligation and inhibited IL-12(p40) mRNA induction by LPS. Finally, bone marrow-derived macrophages from FcR gamma chain-deficient mice, which fail to flux calcium after receptor ligation, failed to inhibit IL-12(p40) mRNA induction. These results indicate that the calcium influxes that occur as a result of receptor ligation are responsible for inhibiting the induction of IL-12 by LPS. Hence, the ligation of phagocytic receptors on macrophages can lead to a dramatic decrease in IL-12 induction. This downregulation may be a way of limiting proinflammatory responses of macrophages to extracellular pathogens, or suppressing the development of cell-mediated immunity to intracellular pathogens.


Assuntos
Interleucina-12/biossíntese , Macrófagos/imunologia , Proteínas de Membrana , Receptores Imunológicos/metabolismo , Receptores de Lipoproteínas , Animais , Anticorpos Monoclonais/imunologia , Cálcio/metabolismo , Cálcio/farmacologia , Regulação para Baixo , Ácido Egtázico/farmacologia , Haemophilus influenzae/imunologia , Interleucina-10/biossíntese , Interleucina-12/genética , Ionomicina/farmacologia , Ligantes , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Complemento/metabolismo , Receptores de IgG/metabolismo , Receptores Depuradores , Receptores Depuradores Classe B , Fator de Necrose Tumoral alfa/biossíntese
3.
J Exp Med ; 189(1): 179-85, 1999 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-9874574

RESUMO

Autoantibodies and immune complexes are major pathogenic factors in autoimmune injury, responsible for initiation of the inflammatory cascade and its resulting tissue damage. This activation results from the interaction of immunoglobulin (Ig)G Fc receptors containing an activation motif (ITAM) with immune complexes (ICs) and cytotoxic autoantibodies which initiates and propagates an inflammatory response. In vitro, this pathway can be interrupted by coligation to FcgammaRIIB, an IgG Fc receptor containing an inhibitory motif (ITIM). In this report, we describe the in vivo consequences of FcgammaRII deficiency in the inflammatory response using a mouse model of IC alveolitis. At subthreshold concentrations of ICs that fail to elicit inflammatory responses in wild-type mice, FcgammaRII-deficient mice developed robust inflammatory responses characterized by increased hemorrhage, edema, and neutrophil infiltration. Bronchoalveolar fluids from FcgammaRII-/- stimulated mice contain higher levels of tumor necrosis factor and chemotactic activity, suggesting that FcgammaRII deficiency lowers the threshold of IC stimulation of resident cells such as the alveolar macrophage. In contrast, complement- and complement receptor-deficient mice develop normal inflammatory responses to suprathreshold levels of ICs, while FcRgamma-/- mice are completely protected from inflammatory injury. An inhibitory role for FcgammaRII on macrophages is demonstrated by analysis of FcgammaRII-/- macrophages which show greater phagocytic and calcium flux responses upon FcgammaRIII engagement. These data reveal contrasting roles for the cellular receptors for IgG on inflammatory cells, providing a regulatory mechanism for setting thresholds for IC sensitivity based on the ratio of ITIM to ITAM FcgammaR expression. Exploiting the FcgammaRII inhibitory pathway could thus provide a new therapeutic approach for modulating antibody-triggered inflammation.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Inflamação/imunologia , Receptores de IgG/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Quimiocinas/metabolismo , Complemento C3/genética , Complemento C3/imunologia , Citocinas/metabolismo , Edema/patologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Transgênicos , Neutrófilos/metabolismo , Fagocitose/imunologia , Alvéolos Pulmonares/patologia , Receptores de IgG/genética
4.
J Exp Med ; 184(6): 2385-92, 1996 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8976192

RESUMO

The role of complement in immunoglobulin G-triggered inflammation was studied in mice genetically deficient in complement components C3 and C4. Using the reverse passive Arthus reaction and experimental models of immune hemolytic anemia and immune thrombocytopenia, we show that these mice have types II and III inflammatory responses that are indistinguishable from those of wild-type animals. Complement-deficient and wild-type animals exhibit comparable levels of erythrophagocytosis and platelet clearance in response to cytotoxic anti-red blood cell and antiplatelet antibodies. Furthermore, in the reverse passive Arthus reaction, soluble immune complexes induce equivalent levels of hemmorhage, edema, and neutrophillic infiltration in complement-deficient and wild-type animals. In contrast, mice that are genetically deficient in the expression of Fc receptors exhibit grossly diminished reactions by both cytotoxic antibodies and soluble immune complexes. These studies provide strong evidence that the activation of cell-based Fc gamma R receptors, but not complement, are required for antibody-triggered murine inflammatory responses.


Assuntos
Anemia Hemolítica/imunologia , Complemento C3/deficiência , Complemento C4/deficiência , Imunoglobulina G/imunologia , Inflamação/imunologia , Trombocitopenia/imunologia , Animais , Plaquetas/imunologia , Complemento C3/imunologia , Complemento C4/imunologia , Eritrócitos/imunologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/fisiologia , Fagocitose , Coelhos
5.
J Exp Med ; 187(4): 641-8, 1998 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-9463414

RESUMO

Coupling of an antibody response to effector cells through the Fc region of antibodies is a fundamental objective of effective vaccination. We have explored the role of the Fc receptor system in a murine model of Cryptococcus neoformans protection by infecting mice deleted for the common gamma chain of FcRs. Passive administration of an IgG1 mAb protects FcRgamma+/- mice infected with C. neoformans, but fails to protect FcRgamma-/- mice, indicating that the gamma chain acting through FcgammaRI and/or III is essential for IgG1-mediated protection. In contrast, passive administration of an IgG3 mAb with identical specificity resulted in enhanced pathogenicity in gamma chain-deficient and wild-type mice. In vitro studies with isolated macrophages demonstrate that IgG1-, IgG2a-, and IgG2b-opsonized C. neoformans are not phagocytosed or arrested in their growth in the absence of the FcRgamma chain. In contrast, opsonization of C. neoformans by IgG3 does not require the presence of the gamma chain or of FcRII, and the internalization of IgG3-treated organisms does not arrest fungal growth.


Assuntos
Criptococose/imunologia , Imunoglobulina G/imunologia , Receptores Fc/imunologia , Animais , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo , Células Cultivadas , Técnicas de Cocultura , Cryptococcus neoformans/crescimento & desenvolvimento , Feminino , Imunoglobulina G/classificação , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Receptores Fc/fisiologia , Formação de Roseta
6.
J Exp Med ; 190(11): 1689-96, 1999 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-10587359

RESUMO

To assess the potency of low-affinity anti-red blood cell (RBC) autoantibodies in the induction of anemia, we generated an immunoglobulin (Ig)G2a class-switch variant of a 4C8 IgM anti-mouse RBC autoantibody, and compared its pathogenic potential with that of its IgM isotype and a high-affinity 34-3C IgG2a autoantibody. The RBC-binding activity of the 4C8 IgG2a variant was barely detectable, at least 1,000 times lower than that of its IgM isotype, having a high-binding avidity, and that of the 34-3C IgG2a monoclonal antibody (mAb). This low-affinity feature of the 4C8 mAb was consistent with the lack of detection of opsonized RBCs in the circulating blood from the 4C8 IgG2a-injected mice. However, the 4C8 IgG2a variant was highly pathogenic, as potent as its IgM isotype and the 34-3C IgG2a mAb, due to its capacity to interact with Fc receptors involved in erythrophagocytosis. In addition, our results indicated that the pentameric form of the low-affinity IgM isotype, by promoting the binding and agglutination of RBCs, is critical for its pathogenic activity. Demonstration of the remarkably high pathogenic potency of low-affinity autoantibodies, if combined with appropriate heavy chain effector functions, highlights the critical role of the Ig heavy chain constant regions, but the relatively minor role of autoantigen-binding affinities, in autoimmune hemolytic anemia.


Assuntos
Anemia Hemolítica Autoimune/imunologia , Autoanticorpos/sangue , Eritrócitos/imunologia , Anemia Hemolítica Autoimune/sangue , Animais , Anticorpos Monoclonais , Linhagem Celular , Citometria de Fluxo , Variação Genética , Hemólise , Imunoglobulina G/sangue , Imunoglobulina G/genética , Imunoglobulina M/sangue , Imunoglobulina M/genética , Região de Troca de Imunoglobulinas , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores Fc/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
7.
J Exp Med ; 187(4): 619-29, 1998 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-9463412

RESUMO

The effector functions of CD4+ T lymphocytes are generally thought to be controlled by distinct populations of regulatory T cells and their soluble products. The role of B cells in the regulation of CD4-dependent host responses is less well understood. Hepatic egg granuloma formation and fibrosis in murine schistosomiasis are dependent on CD4+ lymphocytes, and previous studies have implicated CD8+ T cells or cross-regulatory cytokines produced by T helper (Th) lymphocytes as controlling elements of this pathologic process. In this report, we demonstrate that B cell-deficient (muMT) mice exposed to Schistosoma mansoni develop augmented tissue pathology and, more importantly, fail to undergo the spontaneous downmodulation in disease normally observed during late stages of infection. Unexpectedly, B cell deficiency did not significantly alter T cell proliferative response or cause a shift in the Th1/Th2 balance. Since schistosome-infected Fc receptor-deficient (FcR gamma chain knockout) mice display the same exacerbated egg pathology as that observed in infected muMT mice, the B cell- dependent regulatory mechanism revealed by these experiments appears to require receptor-mediated cell triggering. Together, the data demonstrate that humoral immune response/FcR interactions can play a major role in negatively controlling inflammatory disease induced by CD4+ T cells.


Assuntos
Linfócitos B/fisiologia , Linfócitos T CD4-Positivos/imunologia , Regulação para Baixo , Granuloma/patologia , Receptores Fc/fisiologia , Esquistossomose/patologia , Animais , Linfócitos B/imunologia , Granuloma/imunologia , Granuloma/parasitologia , Fígado/imunologia , Fígado/parasitologia , Fígado/patologia , Hepatopatias/imunologia , Hepatopatias/parasitologia , Hepatopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óvulo/imunologia , Esquistossomose/imunologia , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia
8.
J Cell Biol ; 57(2): 538-50, 1973 May.
Artigo em Inglês | MEDLINE | ID: mdl-4572295

RESUMO

Cytological preparations of Drosophila polytene chromosomes serve as templates for RNA synthesis carried out by exogenous RNA polymerase (Escherichia coli). Incorporation of labeled ribonucleoside triphosphates into RNA may be observed directly by autoradiography. Because of the effects of rifampicin, actinomycin D, ribonuclease, high salt, and the requirement for all four nucleoside triphosphates, we conclude that the labeling observed over chromosomes is due to DNA-dependent RNA polymerase activity. Using this method, one can observe RNA synthesis in vitro on specific chromosome regions due to the activity of exogenous RNA polymerase. We find that much of the RNA synthesis in this system occurs on DNA sequences which appear to be in a nondenatured state.


Assuntos
Cromossomos/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , RNA/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Autorradiografia , Bovinos , Cricetinae , Nucleotídeos de Citosina/metabolismo , RNA Polimerases Dirigidas por DNA/isolamento & purificação , Drosophila , Escherichia coli , Guanosina Trifosfato/metabolismo , Células HeLa , Larva , Filtros Microporos , Hibridização de Ácido Nucleico , Cloreto de Potássio/farmacologia , Ribonucleases/farmacologia , Rifampina/farmacologia , Glândulas Salivares , Moldes Genéticos , Transcrição Gênica/efeitos dos fármacos , Nucleotídeos de Uracila/metabolismo
9.
Science ; 279(5353): 1052-4, 1998 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-9461440

RESUMO

The generation of autoantibody and subsequent tissue deposition of immune complexes (IC) is thought to trigger the pathogenic consequences of systemic autoimmune disease. Modulation of the autoantibody response disrupts pathogenesis by preventing the formation of ICs; however, uncoupling IC formation from subsequent inflammatory responses seems unlikely because of the apparent complexity of the IC-triggered inflammatory cascade. However, the disruption of a single gene, which encodes the gamma chain of the Fc receptor, was found to achieve this uncoupling in a spontaneous model of lupus nephritis, the New Zealand Black/New Zealand White (NZB/NZW) mouse. Gamma chain-deficient NZB/NZW mice generated and deposited IC and activated complement, but were protected from severe nephritis, thus defining another potential pathway for therapeutic intervention in autoimmune disease.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Ativação do Complemento , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Nefrite Lúpica/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Antinucleares/sangue , Complexo Antígeno-Anticorpo/sangue , Proteínas do Sistema Complemento/análise , Cruzamentos Genéticos , Modelos Animais de Doenças , Mesângio Glomerular/imunologia , Mesângio Glomerular/patologia , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Receptores de IgG/genética
10.
J Invest Dermatol ; 138(7): 1539-1545, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29452121

RESUMO

Alopecia areata (AA) is a common autoimmune disease with a lifetime risk of ∼2%. In AA, the immune system targets the hair follicle, resulting in clinical hair loss. The prognosis of AA is unpredictable, and currently there is no definitive treatment. Our previous whole genome expression studies identified active immune circuits in AA lesions, including common γ-chain cytokine and IFN pathways. Because these pathways are mediated through JAK kinases, we prioritized clinical exploration of small molecule JAK inhibitors. In preclinical trials in mice, tofacitinib successfully prevented AA development and reversed established disease. In our tofacitinib trial in 12 patients with moderate to severe AA, 11 patients completed a full course of treatment with minimal adverse events. Following limited response to the initial dose (5 mg b.i.d.), the dose was escalated (10 mg b.i.d.) for nonresponding subjects. Eight of 12 patients demonstrated ≥50% hair regrowth, while three patients demonstrated <50% hair regrowth, as measured by Severity in Alopecia Tool scoring. One patient demonstrated no regrowth. Gene expression profiles and Alopecia Areata Disease Activity Index scores correlated with clinical response. Our open-label studies of ruxolitinib and tofacitinib have shown dramatic clinical responses in moderate to severe AA, providing strong rationale for larger clinical trials using JAK inhibitors in AA. ClinicalTrials.gov ID NCT02299297.


Assuntos
Alopecia em Áreas/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Inibidores de Janus Quinases/uso terapêutico , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Adulto , Alopecia em Áreas/diagnóstico por imagem , Alopecia em Áreas/imunologia , Alopecia em Áreas/patologia , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/imunologia , Biópsia , Relação Dose-Resposta a Droga , Feminino , Perfilação da Expressão Gênica , Folículo Piloso/efeitos dos fármacos , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/patologia , Humanos , Inibidores de Janus Quinases/farmacologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/imunologia , Masculino , Pessoa de Meia-Idade , Nitrilas , Fotografação , Projetos Piloto , Piperidinas/farmacologia , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Pirimidinas/farmacologia , Pirróis/farmacologia , Índice de Gravidade de Doença , Resultado do Tratamento , Adulto Jovem
11.
J Clin Invest ; 102(6): 1258-64, 1998 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9739060

RESUMO

The immune system can recognize self antigens expressed by cancer cells. Differentiation antigens are prototypes of these self antigens, being expressed by cancer cells and their normal cell counterparts. The tyrosinase family proteins are well characterized differentiation antigens recognized by antibodies and T cells of patients with melanoma. However, immune tolerance may prevent immunity directed against these antigens. Immunity to the brown locus protein, gp75/ tyrosinase-related protein-1, was investigated in a syngeneic mouse model. C57BL/6 mice, which are tolerant to gp75, generated autoantibodies against gp75 after immunization with DNA encoding human gp75 but not syngeneic mouse gp75. Priming with human gp75 DNA broke tolerance to mouse gp75. Immunity against mouse gp75 provided significant tumor protection. Manifestations of autoimmunity were observed, characterized by coat depigmentation. Rejection of tumor challenge required CD4(+) and NK1.1(+) cells and Fc receptor gamma-chain, but depigmentation did not require these components. Thus, immunization with homologous DNA broke tolerance against mouse gp75, possibly by providing help from CD4(+) T cells. Mechanisms required for tumor protection were not necessary for autoimmunity, demonstrating that tumor immunity can be uncoupled from autoimmune manifestations.


Assuntos
Vacinas Anticâncer/uso terapêutico , DNA de Neoplasias/uso terapêutico , Melanoma Experimental/prevenção & controle , Glicoproteínas de Membrana , Oxirredutases , Proteínas/uso terapêutico , Vacinação , Vacinas de DNA/uso terapêutico , Animais , Antígenos/imunologia , Antígenos Ly , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/uso terapêutico , Antígenos de Superfície , Autoanticorpos/sangue , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/imunologia , DNA de Neoplasias/imunologia , Cor de Cabelo/genética , Cor de Cabelo/imunologia , Humanos , Tolerância Imunológica , Células Matadoras Naturais/imunologia , Lectinas Tipo C , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subfamília B de Receptores Semelhantes a Lectina de Células NK , Proteínas/genética , Proteínas/imunologia , Receptores de IgG/imunologia , Vacinas de DNA/imunologia
14.
Annu Rev Immunol ; 16: 421-32, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9597136

RESUMO

Recent results obtained in mice deficient in either FcRs or complement have revealed distinct functions for these two classes of molecules. While each is capable of interacting with antibodies or immune complexes, the two systems mediate distinct biological effector responses. Complement-deficient mice are unable to mediate innate immune responses to several bacterial pathogens and bacterial toxins, yet respond normally to the presence of cytotoxic antibodies and pathogenic immune complexes. In contrast, FcR-deficient mice display no defects in innate immunity or susceptibility to a variety of pathogens, yet they are unable to mediate inflammatory responses to cytotoxic IgG antibodies or IgG immune complexes, despite the presence of a normal complement system. These results lead to the surprising conclusion that these two systems have evolved distinct functions in host immunity, with complement and its receptors mediating the interaction of natural antibodies (IgM) with pathogens to effect protection, while FcRs couple the interaction of IgG antibodies to effector cells to trigger inflammatory sequelae. These results necessitate a fundamental revision of the role of these antibody-binding systems in the immune response.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Receptores Fc/fisiologia , Animais , Humanos , Camundongos
15.
Immunity ; 3(1): 21-6, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7621075

RESUMO

Pathogenic self-reactive antibodies are a significant cause of morbidity and mortality and contribute to both cytotoxic and immune complex-triggered inflammatory disorders, typified by rheumatic diseases, autoimmune hemolytic anemia, and thrombocytopenia. Roles have been proposed for Fc receptors, complement, and complement receptors in the pathogenesis of these disorders, although the contribution of each to autoimmune injury is unclear. gamma chain-deficient mice lacking Fc gamma RI and Fc gamma RIII are resistant to the development of experimental immune hemolytic anemia induced by polyclonal rabbit anti-mouse red blood cell IgG antibodies. This resistance is primarily a consequence of ineffective erythrophagocytosis, resulting from the lack of Fc gamma Rs on mononuclear phagocytes. Similarly, gamma chain-deficient mice are completely resistant to the development of experimental immune thrombocytopenia induced by mouse anti-platelet antibodies. These data suggest that Fc receptors play an integral role in the pathogenesis of type II hypersensitivity and suggest potential therapeutic benefits of Fc receptor blockade.


Assuntos
Anemia Hemolítica/imunologia , Citotoxicidade Imunológica , Receptores Fc/imunologia , Trombocitopenia/imunologia , Animais , Anticorpos/administração & dosagem , Eritrócitos/imunologia , Fígado/imunologia , Macrófagos/imunologia , Camundongos , Baço/imunologia
16.
J Immunol ; 161(4): 1908-12, 1998 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9712060

RESUMO

The mechanisms by which Abs mediate protection during blood-stage malaria infections is controversial, with some evidence pointing to the direct effect of Abs on parasite invasion and growth, while other studies suggest that Abs act in cooperation with monocytes to achieve parasite inhibition. To determine whether the effector phase of protection in vivo to the rodent parasite Plasmodium yoelii yoelii requires Fc receptor bearing cells, we passively transferred immune sera into FcR gamma-chain knockout mice. Inflammatory macrophages from these knockout mice were unable to mediate phagocytosis or Ab-dependent cell-mediated cytotoxicity (ADCC) through Fc gamma RI, Fc gamma RII, or Fc gamma RIII. Passive transfer of either P. y. yoelii hyperimmune sera or anti-GST-PYC2 sera directed to the major merozoite surface protein (MSP-1) of this parasite enabled both BALB/cByJ mice and FcR gamma-chain-deficient mice to resist lethal P. y. yoelii 17XL (Py17XL) challenge. mAb302, a protective IgG3 Ab, also passively protected both strains of mice. Most of these samples contain Ab isotypes that would not be able to protect mice if their protective effects required Ab-dependent cell-mediated cytotoxicity. These results establish that, in this infection, protection is directly mediated by Abs and does not require the participation of Fc receptors.


Assuntos
Anticorpos Antiprotozoários/fisiologia , Malária/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Receptores de IgG/fisiologia , Animais , Anticorpos Antiprotozoários/sangue , Especificidade de Anticorpos , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Modelos Animais de Doenças , Glutationa Transferase/genética , Glutationa Transferase/imunologia , Soros Imunes/biossíntese , Imunoglobulina G/sangue , Isotipos de Imunoglobulinas/sangue , Malária/mortalidade , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Plasmodium yoelii/patogenicidade , Receptores de IgG/sangue , Receptores de IgG/genética , Proteínas Recombinantes de Fusão/imunologia
17.
Eur J Immunol ; 29(1): 30-7, 1999 01.
Artigo em Inglês | MEDLINE | ID: mdl-9933083

RESUMO

Following subcutaneous implantation, the murine lymphoma E.G7 [a variant of EL-4, transfected with the chicken ovalbumin (OVA) gene] up-regulates the CD4 molecule. We previously showed that the administration of an anti-CD4 monoclonal antibody (mAb) to EG.7-bearing mice leads to a rapid and complete regression of large established tumors. This tumor regression was shown to require both CD8+ cells and functional Fcgamma receptors (FcgammaR), as it failed to occur in mice depleted of CD8 cells, or mice genetically deficient in FcgammaI/III (gamma-/-mice). Using adoptive transfer, we now show that the FcgammaR+ cells required for this regression are the CD11b+ (phagocytic) cells. Furthermore, experiments using peptide tolerization demonstrated that the critical CD8 CTL population in this model is tumor specific. Analysis of tumors at various stages of regression revealed a massive CD11b+FcgammaR+ and a marginal CD8 infiltration. In the presence of the CTL determinant OVA-8 on tumor cells and of the antitumor mAb, this CD8 infiltration became remarkable, and correlated with tumor regression. These results identify the specific cellular effectors essential for the mAb-mediated tumor regression, and suggest that FcgammaR-activated macrophages induced an expansion of tumor-eliminating CTL in situ.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfoma/imunologia , Linfoma/terapia , Animais , Antígenos CD4/metabolismo , Linfócitos T CD8-Positivos/imunologia , Galinhas , Células-Tronco Hematopoéticas/imunologia , Cinética , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Linfoma/patologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Knockout , Ovalbumina/genética , Ovalbumina/imunologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Linfócitos T Citotóxicos/imunologia , Transfecção
18.
Cell ; 76(3): 519-29, 1994 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-8313472

RESUMO

The gamma subunit of immunoglobulin Fc receptors is an essential component of the high-affinity receptor for IgG (Fc gamma RIII) and is associated with the high-affinity receptor for IgG (Fc gamma RI) and the T cell receptor-CD3 complex. It is required for both receptor assembly and signal transduction. Targeted disruption of this subunit results in immunocompromised mice. Activated macrophages from gamma chain-deficient mice unexpectedly lack the ability to phagocytose antibody-coated particles, despite normal binding. Defects in NK cell-mediated antibody-dependent cytotoxicity and mast cell-mediated allergic responses are evident in these animals, establishing the indispensable role of FcRs in these responses. However, loss of gamma chain does not appear to perturb T cell development, since both thymic and peripheral T cell populations appear normal. These mice thus represent an important tool for evaluating the role of these receptors in humoral and cellular immune responses.


Assuntos
Citotoxicidade Imunológica , Deleção de Genes , Macrófagos Peritoneais/imunologia , Mastócitos/imunologia , Receptores de IgE/fisiologia , Receptores de IgG/fisiologia , Linfócitos T/imunologia , Animais , Sequência de Bases , Southern Blotting , Células Cultivadas , Quimera , DNA/genética , DNA/isolamento & purificação , Primers do DNA , Feminino , Citometria de Fluxo , Genótipo , Interleucina-4/biossíntese , Substâncias Macromoleculares , Masculino , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Anafilaxia Cutânea Passiva , Fagocitose , Reação em Cadeia da Polimerase , Prostaglandina D2/metabolismo , RNA Mensageiro/biossíntese , Receptores de IgE/genética , Receptores de IgG/genética , Baço/imunologia , Células-Tronco
19.
Proc Natl Acad Sci U S A ; 95(2): 652-6, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9435247

RESUMO

Effective tumor immunity requires recognition of tumor cells coupled with the activation of host effector responses. Fc receptor (FcR) gamma-/- mice, which lack the activating Fc gamma R types I and III, did not demonstrate protective tumor immunity in models of passive and active immunization against a relevant tumor differentiation antigen, the brown locus protein gp75. In wild-type mice, passive immunization with mAb against gp75 or active immunization against gp75 prevented the development of lung metastases. This protective response was completely abolished in FcR gamma-deficient mice. Immune responses were intact in gamma-/- mice because IgG titers against gp75 develop normally in gamma-/- mice immunized with gp75. However, uncoupling of the Fc gamma R effector pathway from antibody recognition of tumor antigens resulted in a loss of protection against tumor challenge. These data demonstrate an unexpected and critical role for FcRs in mediating tumor cytotoxicity in vivo and suggest that enhancement of Fc gamma R-mediated antibody-dependent cellular cytotoxicity by inflammatory cells is a key step in the development of effective tumor immunotherapeutics.


Assuntos
Antígenos de Neoplasias/imunologia , Imunidade , Melanoma Experimental/imunologia , Glicoproteínas de Membrana , Oxirredutases , Proteínas/imunologia , Receptores Fc/imunologia , Animais , Anticorpos Monoclonais/imunologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos C57BL , Vacinação
20.
Eur J Immunol ; 27(2): 374-82, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9045907

RESUMO

We investigated the effector mechanisms operating during the rejection of a transplantable solid lymphoma E.G7 (H-2b) which expresses the gene encoding chicken ovalbumin (OVA). Anti-OVA cytotoxic T lymphocytes (CTL) completely and specifically protected animals from the onset of, but could not eradicate established, E.G7 tumors. The growth of the same lymphoma was also effectively prevented by the antibody GK1.5, whose target molecule, CD4, was expressed on E.G7 cells in vivo. Furthermore, GK1.5 was able to eradicate established solid E.G7 tumors. GK1.5-mediated tumor elimination was due to its antitumor activity, and not to the elimination of regulatory CD4+ cells, based on unimpaired tumor growth in the absence of GK1.5 in animals that genetically lack CD4 T cells. In vitro, GK1.5 did not kill tumor cells: complement activation or apoptosis induction were not evident. In vivo, GK1.5-mediated tumor regression did not depend on natural killer cells, but it absolutely required CD8+ cells and intact Fcgamma receptor. We conclude that, in the E.G7 model, the collaboration of antibody and CTL immunity was crucial for the successful immunotherapy of established tumors. The mechanism of this collaboration is discussed.


Assuntos
Anticorpos Antineoplásicos/imunologia , Antígenos CD8/análise , Citotoxicidade Imunológica/imunologia , Rejeição de Enxerto/imunologia , Linfoma/imunologia , Transplante de Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD4/imunologia , Feminino , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/análise , Ovalbumina/imunologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA