Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 138(7): 1539-1545, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29452121

RESUMEN

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.


Asunto(s)
Alopecia Areata/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Inhibidores de las Cinasas Janus/uso terapéutico , Piperidinas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Adulto , Alopecia Areata/diagnóstico por imagen , Alopecia Areata/inmunología , Alopecia Areata/patología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/inmunología , Biopsia , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Folículo Piloso/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/patología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/inmunología , Masculino , Persona de Mediana Edad , Nitrilos , Fotograbar , Proyectos Piloto , Piperidinas/farmacología , Pirazoles/farmacología , Pirazoles/uso terapéutico , Pirimidinas/farmacología , Pirroles/farmacología , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
2.
Immunity ; 14(3): 277-89, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11290337

RESUMEN

Abnormalities of chromosome 1q21 are common in B cell malignancies, but their target genes are largely unknown. By cloning the breakpoints of a (1;14) (q21;q32) chromosomal translocation in a myeloma cell line, we have identified two novel genes, IRTA1 and IRTA2, encoding cell surface receptors homologous to the Fc and inhibitory receptor families. Both genes are selectively expressed in mature B cells: IRTA1 in marginal zone B cells and IRTA2 in centrocytes, marginal zone B cells, and immunoblasts. As a result of the t(1;14), IRTA1 is fused to the immunoglobulin Calpha domain to produce a chimeric IRTA1/Calpha fusion protein. In tumor cell lines with 1q21 abnormalities, IRTA2 expression is deregulated. Thus, IRTA1 and IRTA2 are novel immunoreceptors implicated in B cell development and lymphomagenesis.


Asunto(s)
Linfocitos B/metabolismo , Cromosomas Humanos Par 1/genética , Inmunoglobulinas/química , Linfoma de Células B/genética , Receptores de Superficie Celular/metabolismo , Translocación Genética/genética , Secuencia de Aminoácidos , Linfocitos B/química , Linfocitos B/citología , Linfocitos B/patología , Secuencia de Bases , Rotura Cromosómica/genética , Cromosomas Humanos Par 14/genética , Clonación Molecular , Exones/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal/genética , Humanos , Intrones/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Datos de Secuencia Molecular , Familia de Multigenes/genética , Proteínas de Mieloma/química , Proteínas de Mieloma/genética , Proteínas de Mieloma/metabolismo , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/análisis , ARN Mensajero/genética , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores Fc/química , Células Tumorales Cultivadas
3.
Nat Med ; 6(4): 443-6, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10742152

RESUMEN

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.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/inmunología , Linfoma de Células B/inmunología , Melanoma Experimental/inmunología , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Antígenos CD/genética , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Femenino , Humanos , Inmunización Pasiva , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores de IgG/genética , Rituximab , Trastuzumab
4.
J Exp Med ; 190(11): 1689-96, 1999 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-10587359

RESUMEN

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.


Asunto(s)
Anemia Hemolítica Autoinmune/inmunología , Autoanticuerpos/sangre , Eritrocitos/inmunología , Anemia Hemolítica Autoinmune/sangre , Animales , Anticuerpos Monoclonales , Línea Celular , Citometría de Flujo , Variación Genética , Hemólisis , Inmunoglobulina G/sangre , Inmunoglobulina G/genética , Inmunoglobulina M/sangre , Inmunoglobulina M/genética , Región de Cambio de la Inmunoglobulina , Hígado/inmunología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Receptores Fc/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
5.
Eur J Immunol ; 29(1): 30-7, 1999 01.
Artículo en Inglés | MEDLINE | ID: mdl-9933083

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfoma/inmunología , Linfoma/terapia , Animales , Antígenos CD4/metabolismo , Linfocitos T CD8-positivos/inmunología , Pollos , Células Madre Hematopoyéticas/inmunología , Cinética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Linfoma/patología , Antígeno de Macrófago-1/metabolismo , Ratones , Ratones Noqueados , Ovalbúmina/genética , Ovalbúmina/inmunología , Receptores de IgG/genética , Receptores de IgG/metabolismo , Linfocitos T Citotóxicos/inmunología , Transfección
6.
J Exp Med ; 189(1): 179-85, 1999 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-9874574

RESUMEN

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.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Inflamación/inmunología , Receptores de IgG/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Quimiocinas/metabolismo , Complemento C3/genética , Complemento C3/inmunología , Citocinas/metabolismo , Edema/patología , Macrófagos Alveolares/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/metabolismo , Fagocitosis/inmunología , Alveolos Pulmonares/patología , Receptores de IgG/genética
7.
J Clin Invest ; 102(6): 1258-64, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9739060

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , ADN de Neoplasias/uso terapéutico , Melanoma Experimental/prevención & control , Glicoproteínas de Membrana , Oxidorreductasas , Proteínas/uso terapéutico , Vacunación , Vacunas de ADN/uso terapéutico , Animales , Antígenos/inmunología , Antígenos Ly , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Antígenos de Superficie , Autoanticuerpos/sangre , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/inmunología , ADN de Neoplasias/inmunología , Color del Cabello/genética , Color del Cabello/inmunología , Humanos , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Lectinas Tipo C , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK , Proteínas/genética , Proteínas/inmunología , Receptores de IgG/inmunología , Vacunas de ADN/inmunología
8.
J Immunol ; 161(4): 1908-12, 1998 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9712060

RESUMEN

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.


Asunto(s)
Anticuerpos Antiprotozoarios/fisiología , Malaria/inmunología , Malaria/prevención & control , Plasmodium yoelii/inmunología , Receptores de IgG/fisiología , Animales , Anticuerpos Antiprotozoarios/sangre , Especificidad de Anticuerpos , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Modelos Animales de Enfermedad , Glutatión Transferasa/genética , Glutatión Transferasa/inmunología , Sueros Inmunes/biosíntesis , Inmunoglobulina G/sangre , Isotipos de Inmunoglobulinas/sangre , Malaria/mortalidad , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Plasmodium yoelii/patogenicidad , Receptores de IgG/sangre , Receptores de IgG/genética , Proteínas Recombinantes de Fusión/inmunología
9.
Annu Rev Immunol ; 16: 421-32, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9597136

RESUMEN

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.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Receptores Fc/fisiología , Animales , Humanos , Ratones
10.
Science ; 279(5353): 1052-4, 1998 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-9461440

RESUMEN

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.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Activación de Complemento , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Nefritis Lúpica/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Antinucleares/sangre , Complejo Antígeno-Anticuerpo/sangre , Proteínas del Sistema Complemento/análisis , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Mesangio Glomerular/inmunología , Mesangio Glomerular/patología , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Receptores de IgG/genética
11.
J Exp Med ; 187(4): 619-29, 1998 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-9463412

RESUMEN

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.


Asunto(s)
Linfocitos B/fisiología , Linfocitos T CD4-Positivos/inmunología , Regulación hacia Abajo , Granuloma/patología , Receptores Fc/fisiología , Esquistosomiasis/patología , Animales , Linfocitos B/inmunología , Granuloma/inmunología , Granuloma/parasitología , Hígado/inmunología , Hígado/parasitología , Hígado/patología , Hepatopatías/inmunología , Hepatopatías/parasitología , Hepatopatías/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óvulo/inmunología , Esquistosomiasis/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología
12.
J Exp Med ; 187(4): 641-8, 1998 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-9463414

RESUMEN

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.


Asunto(s)
Criptococosis/inmunología , Inmunoglobulina G/inmunología , Receptores Fc/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo , Células Cultivadas , Técnicas de Cocultivo , Cryptococcus neoformans/crecimiento & desarrollo , Femenino , Inmunoglobulina G/clasificación , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Fagocitosis , Receptores Fc/fisiología , Formación de Roseta
13.
Proc Natl Acad Sci U S A ; 95(2): 652-6, 1998 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-9435247

RESUMEN

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.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunidad , Melanoma Experimental/inmunología , Glicoproteínas de Membrana , Oxidorreductasas , Proteínas/inmunología , Receptores Fc/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Inmunización Pasiva , Ratones , Ratones Endogámicos C57BL , Vacunación
14.
J Exp Med ; 185(11): 1977-85, 1997 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-9166427

RESUMEN

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.


Asunto(s)
Interleucina-12/biosíntesis , Macrófagos/inmunología , Proteínas de la Membrana , Receptores Inmunológicos/metabolismo , Receptores de Lipoproteína , Animales , Anticuerpos Monoclonales/inmunología , Calcio/metabolismo , Calcio/farmacología , Regulación hacia Abajo , Ácido Egtácico/farmacología , Haemophilus influenzae/inmunología , Interleucina-10/biosíntesis , Interleucina-12/genética , Ionomicina/farmacología , Ligandos , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Complemento/metabolismo , Receptores de IgG/metabolismo , Receptores Depuradores , Receptores Depuradores de Clase B , Factor de Necrosis Tumoral alfa/biosíntesis
15.
Eur J Immunol ; 27(2): 374-82, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9045907

RESUMEN

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.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Antígenos CD8/análisis , Citotoxicidad Inmunológica/inmunología , Rechazo de Injerto/inmunología , Linfoma/inmunología , Trasplante de Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD4/inmunología , Femenino , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/análisis , Ovalbúmina/inmunología , Células Tumorales Cultivadas
16.
J Exp Med ; 184(6): 2385-92, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8976192

RESUMEN

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.


Asunto(s)
Anemia Hemolítica/inmunología , Complemento C3/deficiencia , Complemento C4/deficiencia , Inmunoglobulina G/inmunología , Inflamación/inmunología , Trombocitopenia/inmunología , Animales , Plaquetas/inmunología , Complemento C3/inmunología , Complemento C4/inmunología , Eritrocitos/inmunología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/fisiología , Fagocitosis , Conejos
17.
Immunity ; 3(1): 21-6, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7621075

RESUMEN

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.


Asunto(s)
Anemia Hemolítica/inmunología , Citotoxicidad Inmunológica , Receptores Fc/inmunología , Trombocitopenia/inmunología , Animales , Anticuerpos/administración & dosificación , Eritrocitos/inmunología , Hígado/inmunología , Macrófagos/inmunología , Ratones , Bazo/inmunología
18.
Cell ; 76(3): 519-29, 1994 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-8313472

RESUMEN

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.


Asunto(s)
Citotoxicidad Inmunológica , Eliminación de Gen , Macrófagos Peritoneales/inmunología , Mastocitos/inmunología , Receptores de IgE/fisiología , Receptores de IgG/fisiología , Linfocitos T/inmunología , Animales , Secuencia de Bases , Southern Blotting , Células Cultivadas , Quimera , ADN/genética , ADN/aislamiento & purificación , Cartilla de ADN , Femenino , Citometría de Flujo , Genotipo , Interleucina-4/biosíntesis , Sustancias Macromoleculares , Masculino , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Anafilaxis Cutánea Pasiva , Fagocitosis , Reacción en Cadena de la Polimerasa , Prostaglandina D2/metabolismo , ARN Mensajero/biosíntesis , Receptores de IgE/genética , Receptores de IgG/genética , Bazo/inmunología , Células Madre
19.
Proc Natl Acad Sci U S A ; 85(16): 6067-71, 1988 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-3137564

RESUMEN

A retroviral vector, RIM, containing murine c-myc under the control of immunoglobulin heavy-chain gene promoter and enhancer elements and v-Ha-ras driven by a Moloney murine leukemia virus long terminal repeat induced IgM-secreting plasmacytomas in 28% of adult and 83% of 3-week-old pristane-conditioned mice with mean latency periods of 60-70 days. In contrast, the same vector only harboring c-myc or v-Ha-ras was virtually ineffective. RIM-induced plasmacytomas expressed retroviral myc and ras genes while their endogenous c-myc alleles were unrearranged and transcriptionally inactive. These plasmacytomas were clonal as each possessed a unique immunoglobulin heavy-chain joining region rearrangement and a single recombinant provirus. Moloney murine leukemia helper virus did not play an obligatory role in tumorigenesis since insertions of Moloney murine leukemia proviruses were found in only 6 of 24 plasmacytomas induced in adult mice. Taken together, these findings support the view that the v-Ha-ras oncogene can cooperate with an activated myc gene in pristane plasmacytomagenesis.


Asunto(s)
Elementos de Facilitación Genéticos , Genes de Inmunoglobulinas , Cadenas Pesadas de Inmunoglobulina/metabolismo , Inmunoglobulina M/metabolismo , Plasmacitoma/etiología , Regiones Promotoras Genéticas , Proto-Oncogenes , Retroviridae/genética , Terpenos/farmacología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Plasmacitoma/inmunología , Recombinación Genética
20.
Eur J Immunol ; 18(7): 1101-9, 1988 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2969819

RESUMEN

Injection of newborn mice with mixtures of wild-type moloney murine leukemia (Mo-MuLV) virus and other recombinant retroviruses harboring the myc oncogene alone or in combination with the H-ras oncogene resulted in a 100% incidence of lymphatic leukemias from which permanent cell lines could be established in vitro. These cells are immunoglobulin (Ig)-, Thy-1+BP- and CD8-CD4- indicating that they are early thymocytes. Such transformed pre-T lines lack retroviral myc and ras genes but occasionally possess proviral insertion near to their endogenous myc and pim genes. We show that both Ig heavy chain (Igh) and T cell receptor (TcR) genes are rearranged in most of these lines. In some cases, a primary recombination was followed by a secondary rearrangement at the same locus. We show that VT gamma genes can rearrange outside of their known cluster suggesting that TcR gamma diversification in such pre-T cells may be different to that in more mature T cells. Ig D-JH recombinations may precede TcR gene recombination in these early T cell lines, and some but not all express sterile Cmu transcripts. Some of these lines express surface heterodimers that appear composed of alpha and beta chains that can be immunoprecipitated with a monoclonal anti-T3 antibody but not with the anti-V beta 8 monoclonal antibody F23.1. This established pre-T cell line represents novel biological material for the dissection of T cell development and function analogous to A-MuLV transformed pre-B cells.


Asunto(s)
Genes de Inmunoglobulinas , Cadenas Pesadas de Inmunoglobulina/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/fisiología , Animales , Antígenos de Diferenciación de Linfocitos T/análisis , Diferenciación Celular , Regulación de la Expresión Génica , Ratones , Oncogenes , Receptores de Antígenos de Linfocitos B/análisis , Receptores de Antígenos de Linfocitos T alfa-beta , Receptores de Antígenos de Linfocitos T gamma-delta , Linfocitos T/citología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...