Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Exp Immunol ; 192(1): 104-119, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29178470

RESUMEN

Haemophagocytic lymphohistiocytosis (HLH) constitutes a spectrum of immunological disorders characterized by uncontrolled immune activation and key symptoms such as fever, splenomegaly, pancytopenia, haemophagocytosis, hyperferritinaemia and hepatitis. In genetic or primary HLH, hyperactivated CD8+ T cells are the main drivers of pathology. However, in acquired secondary HLH, the role of lymphocytes remains vague. In the present study the involvement of lymphocytes in the pathogenesis of a cytomegalovirus-induced model of secondary HLH was explored. We have previously reported CD8+ T cells to be redundant in this model, and therefore focused on CD4+ helper and regulatory T cells. CD4+ T cells were activated markedly and skewed towards a proinflammatory T helper type 1 transcription profile in mice displaying a severe and complete HLH phenotype. Counter to expectations, regulatory T cells were not reduced in numbers and were, in fact, more activated. Therapeutic strategies targeting CD25high hyperactivated T cells were ineffective to alleviate disease, indicating that T cell hyperactivation is not a pathogenic factor in cytomegalovirus-induced murine HLH. Moreover, even though T cells were essential in controlling viral proliferation, CD4+ T cells, in addition to CD8+ T cells, were dispensable in the development of the HLH-like syndrome. In fact, no T or B cells were required for induction and propagation of HLH disease, as evidenced by the occurrence of cytomegalovirus-associated HLH in severe combined immunodeficient (SCID) mice. These data suggest that lymphocyte-independent mechanisms can underlie virus-associated secondary HLH, accentuating a clear distinction with primary HLH.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Herpesviridae/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/patología , Linfocitos T Reguladores/inmunología , Animales , Infecciones por Herpesviridae/complicaciones , Interferón gamma/genética , Activación de Linfocitos , Depleción Linfocítica , Linfohistiocitosis Hemofagocítica/virología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Muromegalovirus , Células TH1/inmunología
2.
Ann Rheum Dis ; 68(5): 744-50, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18480308

RESUMEN

OBJECTIVES: Patients with rheumatoid arthritis (RA) have defective CD4(+)CD25(+) regulatory T (T(reg)) cells and increased osteoclastogenesis. A similar situation has been described in collagen-induced arthritis (CIA). In this study, it was investigated whether a single transfer of polyclonally activated T(reg) cells inhibits CIA and osteoclastogenesis. METHODS: Purified T(reg) cells were expanded in vitro with anti-CD3 and anti-CD28 antibody-coated beads and injected into DBA/1 mice. Mice were immunised with collagen type II (CII) in complete Freund adjuvant (CFA) and scores of arthritis were recorded. In vitro osteoclastogenesis assays were performed on splenocytes by stimulation with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor (NF)kappaB ligand (RANKL). Levels of anti-CII antibody and cytokines were determined in the supernatant using ELISA and Bio-Plex protein array system. RESULTS: It was found that 10(6) activated T(reg) cells significantly counteracted the development of CIA, which was accompanied by decreased serum levels of TNFalpha and IL6, but not by inhibition of autoimmune antibody responses. The differentiation of osteoclasts in splenocyte cultures was significantly reduced in the presence of prestimulated T(reg) cells. Expression of cytokines that are described to inhibit osteoclastogenesis, including granulocyte macrophage colony-stimulating factor (GM-CSF), interferon (IFN)gamma, interleukin (IL)5 and IL10, were dramatically increased upon addition of T(reg) cells. Furthermore, splenocytes from mice that had been treated with T(reg) cells displayed an impaired capacity to develop into mature osteoclasts, suggesting that T(reg) cells abrogated osteoclastogenesis in vivo. CONCLUSIONS: Activated CD4(+)CD25(+) T(reg) cells improve clinical symptoms of CIA, regulate cytokine production and inhibit osteoclastogenesis in vitro and in vivo.


Asunto(s)
Artritis Experimental/prevención & control , Osteoclastos/patología , Linfocitos T Reguladores/trasplante , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/análisis , Interleucina-6/biosíntesis , Activación de Linfocitos/inmunología , Transfusión de Linfocitos , Ratones , Ratones Endogámicos DBA , Factor de Necrosis Tumoral alfa/biosíntesis
3.
Clin Exp Immunol ; 130(3): 379-85, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12452826

RESUMEN

TCR/CD3 aggregation by injection of anti-CD3 Ab produces T cell activation, release of cytokines such as IFN-gamma, and apoptosis in the cortical region of the thymus. We show that anti-CD3 Ab induces IL-15 mRNA in spleens of wild-type but not IFN-gamma receptor-knock-out (IFN-gammaR KO) mice. The loss of IL-15 mRNA induction in IFN-gammaR KO mice was associated with increased thymocyte apoptosis. Pretreatment of wild-type mice with neutralizing anti-IL-15 Ab increased the anti-CD3-triggered thymocyte apoptosis, thus mimicking the sensitive phenotype of IFN-gammaR KO mice. Inversely, anti-CD3-induced apoptosis in IFN-gammaR KO mice was suppressed by administration of recombinant IL-15. In IFN-gammaR KO mice and in wild-type mice that were treated with anti-IL-15, augmented apoptosis affected mainly CD4+CD8+ immature thymocytes. IL-15 as well as IL-15Ralpha mRNA expression in thymocytes was not increased by anti-CD3. These data demonstrate that systemic IL-15 exerts anti-apoptotic activity on immature T cells and establish a regulatory mechanism whereby TCR/CD3 engagement induces IL-15 expression via an IFN-gamma-dependent pathway. The self-amplifying nature of this IFN-gamma/IL-15 connection may constitute a regulatory pathway in central tolerance to self.


Asunto(s)
Interferón gamma/inmunología , Interleucina-15/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Autotolerancia , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis , Complejo CD3/inmunología , Citometría de Flujo , Interferón gamma/genética , Activación de Linfocitos , Ratones , Ratones Noqueados
4.
J Immunol ; 163(6): 3503-10, 1999 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10477624

RESUMEN

Induction of experimental autoimmune diseases often relies on immunization with the organ-specific autoantigens in CFA, which contains heat-killed mycobacteria. In several of these models, including collagen-induced arthritis, endogenous IFN-gamma acts as a disease-limiting factor in the pathogenesis of the disease. Here we show that in collagen-induced arthritis the protective effect of IFN-gamma depends on the presence of mycobacteria in the adjuvant. Omission of mycobacteria inverts the role of endogenous IFN-gamma to a disease-promoting factor. Thus, the mycobacterial component of CFA opens a pathway by which endogenous IFN-gamma exerts a protective effect that supersedes its otherwise disease-promoting effect. Extramedullary hemopoiesis and expansion of the Mac-1+ cell population accompanied the accelerated and more severe disease course in the IFN-gamma receptor knockout mice immunized with CFA. Treatment of such mice with Abs against the myelopoietic cytokines IL-6 or IL-12 inhibited both disease development and the expansion of the Mac-1+ population. We postulate that mycobacteria in CFA stimulate the expansion of the Mac-1+ cell population by a hemopoietic process that is restrained by endogenous IFN-gamma. These results have important implications for the validity of animal models of autoimmunity to study the pathogenesis and to evaluate cytokine-based therapy of autoimmune diseases.


Asunto(s)
Artritis Experimental/genética , Artritis Experimental/inmunología , Enfermedades Autoinmunes/genética , Adyuvante de Freund/inmunología , Células Madre Hematopoyéticas/inmunología , Antígeno de Macrófago-1/biosíntesis , Mycobacterium/inmunología , Receptores de Interferón/genética , Animales , Artritis Experimental/patología , Artritis Experimental/prevención & control , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/prevención & control , Colágeno/inmunología , Femenino , Adyuvante de Freund/administración & dosificación , Eliminación de Gen , Predisposición Genética a la Enfermedad , Hematopoyesis Extramedular/genética , Hematopoyesis Extramedular/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Hipersensibilidad Tardía/genética , Hipersensibilidad Tardía/inmunología , Sueros Inmunes/farmacología , Esquemas de Inmunización , Interferón gamma/fisiología , Interleucina-12/inmunología , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Receptores de Interferón/deficiencia , Bazo/inmunología , Bazo/patología , Receptor de Interferón gamma
5.
Eur J Immunol ; 28(7): 2143-51, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9692883

RESUMEN

In several models of inflammation, including collagen-induced arthritis (CIA), the disease-promoting effect of IL-12 has been attributed to its well-known ability to produce IFN-gamma. However, IFN-gamma receptor knockout (IFN-gammaR KO) mice of the DBA/1 strain have been reported to be more susceptible to CIA than corresponding wild-type mice, indicating the existence of an IFN-gamma-mediated protective pathway in this model. In the present study the development of CIA was found to be completely prevented by pretreatment with a neutralizing anti-IL-12 antibody, not only in wild-type, but significantly also in IFN-gammaR KO mice. In both strains of mice, the protective effect of anti-IL-12 was associated with lower production of anti-collagen type II antibodies. In vivo stimulation with anti-CD3 antibody in arthritic IFN-gammaR KO mice resulted in production of higher levels of circulating IFN-gamma, TNF and IL-2 than in corresponding control mice that had not received the arthritis-inducing immunization. This was not the case in arthritis-developing wild-type mice. Furthermore, the protective effect of anti-IL-12 antibody in mutant, but not in wild-type mice, was associated with lower circulating IFN-gamma, TNF and IL-2 and higher IL-4 and IL-5 cytokine levels following an anti-CD3 challenge. The data indicate that IL-12 promotes the development of arthritis independently of its ability to induce or favor production of IFN-gamma. In fact, any IFN-gamma produced in the course of the disease process rather exerts a protective effect. Furthermore, our study suggests that, in the absence of a functional IFN-gamma system, endogenous IL-12 exerts its disease-promoting effect by favoring production of other Th1-associated cytokines (IL-2 and TNF), by inhibiting development of IL-4- and IL-5-producing T cells and by stimulating production of anti-collagen autoantibodies.


Asunto(s)
Artritis/prevención & control , Colágeno/inmunología , Interleucina-12/fisiología , Receptores de Interferón/fisiología , Animales , Citocinas/biosíntesis , Inmunoglobulina G/biosíntesis , Interferón gamma/fisiología , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptor de Interferón gamma
6.
Eur J Biochem ; 243(3): 762-9, 1997 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9057843

RESUMEN

Human osteosarcoma cells secrete a novel C-X-C chemokine called granulocyte chemotactic protein-2 (GCP-2), which was previously identified by amino acid sequencing of the purified natural protein. In order to understand the role of this new protein in inflammatory reactions, we cloned GCP-2 DNA sequences to generate recombinant protein and specific DNA probes and primers. By means of PCR on cloned cDNA of osteosarcoma cells induced by interleukin-1 beta and fibroblasts induced by lipopolysaccharide plus dsRNA, the complete coding domain of GCP-2 was isolated. This sequence was cloned into the bacterial expression vector pHEN1 and, after induction, GCP-2 was secreted into the periplasm of Escherichia coli. Recombinant GCP-2 (rGCP-2) was purified and characterized by SDS/PAGE as a monomeric 6.5-kDa protein and by amino-terminal sequencing. The chemoattractive potency of GCP-2 for neutrophilic granulocytes was about 10-times less than that of interleukin-8 and the minimal effective dose was 10 ng/ml. However, at optimal dose (100 ng/ml) the maximal chemotactic response was comparable with that of interleukin-8. Both characteristics correspond with those of natural GCP-2. In addition, intracellular calcium release in neutrophils by recombinant GCP-2 was achieved with as little as 10 ng/ml. Quantitation studies using reverse transcriptase and the polymerase chain reaction revealed higher GCP-2 mRNA production in normal fibroblasts than in tumor cells. When compared with epithelial-cell-derived neutrophil-activating peptide-78 (ENA-78) mRNA, the GCP-2 mRNA levels were higher in all cell lines tested. In addition, GCP-2 and ENA-78 expression seem to be differentially regulated in that phorbol ester and lipopolysaccharide have opposing effects on their mRNA induction in diploid fibroblasts and epithelial cells, respectively. Interleukin-1 was demonstrated to be a general inducer for both chemokines, while interferon-gamma down-regulates their mRNA expression. The availability of recombinant GCP-2 together with the quantitation studies on mRNA expression will help to further elucidate the biological role of GCP-2 during the inflammatory response.


Asunto(s)
Quimiocinas CXC , Quimiocinas/biosíntesis , Quimiocinas/genética , Vectores Genéticos , Interleucina-8/genética , ARN Mensajero/biosíntesis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Secuencia de Aminoácidos , Secuencia de Bases , Quimiocina CXCL5 , Quimiocina CXCL6 , Quimiocinas/química , Quimiocinas/aislamiento & purificación , Quimiotaxis de Leucocito , Clonación Molecular , Codón/aislamiento & purificación , Células Epiteliales , Epitelio/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Granulocitos/fisiología , Humanos , Interleucina-8/análogos & derivados , Datos de Secuencia Molecular , Activación Neutrófila
7.
Infect Immun ; 63(4): 1158-64, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7890366

RESUMEN

Administration of staphylococcal enterotoxin B (SEB) to BALB/c mice was found to induce a cytokine release syndrome hallmarked by weight loss and hypoglycemia. A neutralizing monoclonal antibody against gamma interferon (IFN-gamma) given before SEB counteracted weight loss and prevented hypoglycemia. This protective effect of anti-IFN-gamma antibody was associated with decreased IFN-gamma levels in serum; tumor necrosis factor (TNF) and interleukin-6 (IL-6) levels remained unchanged. A monoclonal anti-IL-6 antibody, known for its ability to cause accumulation of biologically active IL-6 in the circulation, did not modify SEB-induced body weight loss or hypoglycemia. Levels of TNF, IFN-gamma, and IL-6 in serum were all more elevated in anti-IL-6-treated mice than in corresponding SEB-challenged control mice. In D-galactosamine-sensitized mice, SEB-induced weight loss but not hypoglycemia was more severe, resulting mostly in death within 24 h. Higher levels of biologically active TNF and IFN-gamma in serum were noted in these mice than in mice receiving SEB only. In D-galactosamine-sensitized mice, anti-IFN-gamma antibody did prevent hypoglycemia but failed to reduce the severity of the syndrome. Again, TNF levels in anti-IFN-gamma-treated mice remained unchanged. Pretreatment with anti-IL-6 antibody temporarily attenuated SEB-induced hypoglycemia in sensitized mice. Thus, at 6 h post-SEB injection, anti-IL-6-treated mice were less hypoglycemic than corresponding controls. However, at 24 h, hypoglycemia was significantly aggravated. Concomitantly, IL-6 levels were dramatically increased. Neither anti-IFN-gamma nor anti-IL-6 antibody treatment modulated mortality levels in D-galactosamine-sensitized mice. The data obtained with anti-IFN-gamma antibody clearly indicate that endogenous IFN-gamma is instrumental in bringing about hypoglycemia and body weight loss in mice exposed to SEB but also that hypoglycemia is not a crucial determinant of mortality in D-galactosamine-sensitized mice. The data obtained with anti-IL-6 antibody indicate that endogenous IL-6 is involved in regulating the levels of TNF and IFN-gamma in serum.


Asunto(s)
Citocinas/metabolismo , Enterotoxinas/farmacología , Interferón gamma/fisiología , Interleucina-6/fisiología , Superantígenos/farmacología , Animales , Peso Corporal , Diarrea/inducido químicamente , Galactosamina/toxicidad , Hipoglucemia/fisiopatología , Técnicas Inmunológicas , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...