Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 11(3): e0151252, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26978520

RESUMEN

The precise context in which the innate immune system is activated plays a pivotal role in the subsequent instruction of CD4+ T helper (Th) cell responses. Th1 responses are downregulated when antigen is encountered in the presence of antigen-IgG immune complexes. To assess if Th17 responses to antigen are subject to similar influences in the presence of immune complexes we utilized an inflammatory airway disease model in which immunization of mice with Complete Freund's Adjuvant (CFA) and ovalbumin (Ova) induces a powerful Ova-specific Th1 and Th17 response. Here we show that modification of that immunization with CFA to include IgG-Ova immune complexes results in the suppression of CFA-induced Th17 responses and a concurrent enhancement of Ova-specific Th2 responses. Furthermore, we show the mechanism by which these immune complexes suppress Th17 responses is through the enhancement of IL-10 production. In addition, the generation of Th17 responses following immunization with CFA and Ova were dependent on IL-1α but independent of NLRP3 inflammasome activation. Together these data represent a novel mechanism by which the generation of Th17 responses is regulated.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Inflamasomas/inmunología , Células Th17/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Adyuvante de Freund , Inmunización , Inflamasomas/metabolismo , Interleucina-1alfa/metabolismo , Ratones , Ovalbúmina , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
2.
J Immunol ; 193(10): 5190-8, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25320279

RESUMEN

IgG immune complexes have been shown to modify immune responses driven by APCs in either a pro- or anti-inflammatory direction depending upon the context of stimulation. However, the ability of immune complexes to modulate the inflammasome-dependent innate immune response is unknown. In this study, we show that IgG immune complexes suppress IL-1α and IL-1ß secretion through inhibition of inflammasome activation. The mechanism by which this inhibition occurs is via immune complex ligation of activating FcγRs, resulting in prevention of both activation and assembly of the inflammasome complex in response to nucleotide-binding domain leucine-rich repeat (NLR) P3, NLRC4, or AIM2 agonists. In vivo, administration of Ag in the form of an immune complex during priming of the immune response inhibited resultant adaptive immune responses in an NLRP3-dependent model of allergic airway disease. Our data reveal an unexpected mechanism regulating CD4(+) T cell differentiation, by which immune complexes suppress inflammasome activation and the generation of IL-1α and IL-1ß from APCs, which are critical for the Ag-driven differentiation of CD4(+) T cells.


Asunto(s)
Complejo Antígeno-Anticuerpo/genética , Linfocitos T CD4-Positivos/inmunología , Inflamasomas/inmunología , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pulmón/inmunología , Hipersensibilidad Respiratoria/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Compuestos de Alumbre/administración & dosificación , Animales , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/patología , Regulación de la Expresión Génica , Inmunidad Innata , Inflamasomas/genética , Interleucina-1alfa/biosíntesis , Interleucina-1beta/biosíntesis , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina , Receptores de IgG/genética , Receptores de IgG/inmunología , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/patología , Transducción de Señal
3.
Ann N Y Acad Sci ; 1319: 82-95, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24840700

RESUMEN

Inflammasomes continue to generate interest in an increasing number of disciplines owing to their unique ability to integrate a myriad of signals from pathogen- and damage-associated molecular patterns into a proinflammatory response. This potent caspase-1-dependent process is capable of activating the innate immune system, initiating pyroptosis (an inflammatory form of programmed cell death), and shaping adaptive immunity. The NLRP3 inflammasome is the most thoroughly studied of the inflammasome complexes that have been described thus far, perhaps owing to its disparate assortment of agonists. This review highlights our current understanding of the mechanisms of both priming and activation of the NLRP3 inflammasome.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/fisiología , Inmunidad Adaptativa/inmunología , Animales , Muerte Celular/inmunología , Supervivencia Celular/inmunología , Humanos , Inflamasomas/química , Proteína con Dominio Pirina 3 de la Familia NLR
4.
Arthritis Rheumatol ; 66(4): 852-62, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24757138

RESUMEN

OBJECTIVE: Arthritis and valvular carditis coexist in several human rheumatic diseases, including systemic lupus erythematosus, rheumatic fever, and rheumatoid arthritis. T cell receptor-transgenic K/BxN mice develop spontaneous autoantibody-associated arthritis and valvular carditis. The common Fc receptor γ (FcRγ) signaling chain is required for carditis to develop in K/BxN mice. FcRγ pairs with numerous receptors in a variety of cells. The aim of this study was to identify the FcRγ-associated receptors and Fcγ receptor (FcγR)-expressing cells that mediate valvular carditis in this model. METHODS: We bred K/BxN mice lacking the genes that encode activating Fcγ receptors (FcγRI, FcγRIII, and FcγRIV), and we assessed these mice for valvular carditis. We similarly evaluated complement component C3-deficient K/BxN mice. Immunohistochemistry, bone marrow transplantation, and macrophage depletion were used to define the key FcRγ-expressing cell type. RESULTS: Genetic deficiency of only one of the activating Fcγ receptors did not prevent carditis, whereas deficiency of all 3 activating Fcγ receptors did. Further analysis demonstrated that FcγRIII and FcγRIV were the key drivers of valve inflammation; FcγRI was dispensable. C3 was not required. FcRγ expression by radioresistant cells was critical for valvular carditis to develop, and further analysis indicated that macrophages were the key candidate FcγR-expressing effectors of carditis. CONCLUSION: FcγRIII and FcγRIV act redundantly to promote valvular carditis in K/BxN mice with systemic autoantibody-associated arthritis. Macrophage depletion reduced the severity of valve inflammation. These findings suggest that pathogenic autoantibodies engage Fcγ receptors on macrophages to drive valvular carditis. Our study provides new insight into the pathogenesis of cardiovascular inflammation in the setting of autoantibody-associated chronic inflammatory diseases.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Macrófagos/inmunología , Miocarditis/inmunología , Receptores de IgG/metabolismo , Animales , Artritis Experimental/genética , Artritis Experimental/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Autoanticuerpos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Miocarditis/genética , Miocarditis/metabolismo , Receptores de IgG/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
Eur J Immunol ; 43(8): 2003-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23928964

RESUMEN

The NLRP3 inflammasome plays a critical role in regulating inflammatory and cell death pathways in response to a diverse array of stimuli. Activation of the NLRP3 inflammasome results in activation of the cysteine protease caspase-1 and the subsequent processing and secretion of the proinflammatory cytokines IL-1ß and IL-18. In this issue of the European Journal of Immunology, Licandro et al. [Eur. J. Immunol. 2013. 43, 2126-2137] show that the NLRP3 inflammasome contributes to oxidative DNA damage. In addition, activation of the NLRP3 inflammasome modulates a number of pathways involved in DNA damage repair, cell cycle, and apoptosis, suggesting a novel role for the NLRP3 inflammasome in DNA damage responses following cellular stress.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN/efectos de los fármacos , Reparación del ADN/genética , Células Dendríticas/metabolismo , Inflamasomas/inmunología , Animales , Proteína con Dominio Pirina 3 de la Familia NLR
6.
J Immunol ; 191(3): 1055-62, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23794629

RESUMEN

The class A macrophage scavenger receptor Msr1 (SR-A, CD204) has been reported to participate in the maintenance of immunological tolerance. We investigated the role of Msr1 in a mouse model of autoantibody-dependent arthritis. Genetic deficiency of Msr1 in K/BxN TCR transgenic mice decreased the incidence and severity of arthritis because of decreased autoantibody production. Despite normal initial activation of autoreactive CD4(+) T cells, potentially autoreactive B cells in Msr1(-/-) K/BxN mice retained a naive phenotype and did not expand. This was not due to an intrinsic B cell defect. Rather, we found that macrophages lacking Msr1 were inefficient at taking up the key autoantigen glucose-6-phosphate isomerase and that Msr1-deficient mice had elevated serum concentrations of glucose-6-phosphate isomerase. Arthritis developed normally when bone marrow from Msr1(-/-) K/BxN mice was transplanted into hosts whose macrophages did express Msr1. Thus, Msr1 can regulate the concentration of a soluble autoantigen. In this model, the absence of Msr1 led to higher levels of soluble autoantigen and protected mice from developing pathogenic autoantibodies, likely because of altered cognate interactions of autoreactive T and B cells with impaired differentiation of follicular Th cells.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Autoinmunidad , Linfocitos B/inmunología , Glucosa-6-Fosfato Isomerasa/inmunología , Receptores Depuradores de Clase A/metabolismo , Animales , Artritis Experimental/inmunología , Autoanticuerpos/biosíntesis , Autoantígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Glucosa-6-Fosfato Isomerasa/sangre , Glucosa-6-Fosfato Isomerasa/metabolismo , Activación de Linfocitos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/inmunología
7.
Arthritis Res Ther ; 14(6): R269, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23237573

RESUMEN

INTRODUCTION: The effector functions of immunoglobulin G (IgG) are mediated by interaction of its Fc region with Fc receptors (FcγRs) and/or the complement system. The three main pathways of complement activation converge at C3. However, C3-independent pathways can activate C5 and other downstream complement components during IgG-initiated inflammatory responses. These C3-independent pathways of C5 activation are triggered by activating FcγRs in some systems or can be activated by factors of the coagulation cascade such as thrombin. Here we studied the interplay of C3, C5, and activating FcγRs in a model of spontaneous autoantibody-driven arthritis. METHODS: We utilized the K/BxN TCR transgenic mouse model of arthritis. We bred K/BxN mice bearing targeted or naturally-occurring mutations in one or more of the genes encoding complement components C3, C5, and FcRγ, the cytoplasmic signaling chain shared by the activating FcγRs. We measured arthritis development, the production of arthritogenic autoantibodies, T cell activation status and cytokine synthesis. In addition, we treated mice with anti-C5 monoclonal antibodies or with the thrombin inhibitor argatroban. RESULTS: We have previously shown that genetic deficiency of C5 protects K/BxN mice from the development of arthritis. We found here that C3-deficient K/BxN mice developed arthritis equivalent in severity to C3-sufficient animals. Arthritis also developed normally in K/BxN mice lacking both C3 and FcRγ, but could be ameliorated in these animals by treatment with anti-C5 monoclonal antibody or by treatment with argatroban. Production of arthritogenic autoantibodies, T cell activation, and T cell cytokine production were not affected by the absence of C3, C5, and/or FcRγ. CONCLUSIONS: In K/BxN mice, C5-dependent autoantibody-driven arthritis can occur in the genetic absence of both complement C3 and activating FcγRs. Our findings suggest that in this setting, thrombin activates C5 to provoke arthritis.


Asunto(s)
Artritis/inmunología , Autoanticuerpos/inmunología , Complemento C3/inmunología , Complemento C5/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antitrombinas/farmacología , Arginina/análogos & derivados , Artritis/genética , Artritis/metabolismo , Autoanticuerpos/sangre , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/inmunología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C3/deficiencia , Complemento C3/genética , Complemento C5/deficiencia , Complemento C5/genética , Femenino , Citometría de Flujo , Glucosa-6-Fosfato Isomerasa/inmunología , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Ácidos Pipecólicos/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de IgG/deficiencia , Receptores de IgG/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sulfonamidas
8.
Eur J Immunol ; 42(9): 2354-62, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22706882

RESUMEN

Allelic exclusion of antigen receptor loci is a fundamental mechanism of immunological self-tolerance. Incomplete allelic exclusion leads to dual T-cell receptor (TCR) expression and can allow developing autoreactive αß T lymphocytes to escape clonal deletion. Because allelic exclusion at the TCR-ß locus is more stringent than at the TCR-α locus, dual TCR-ß expression has not been considered a likely contributor to autoimmunity. We show here that incomplete TCR-ß allelic exclusion permits developing thymocytes bearing the autoreactive, transgene-encoded KRN TCR to be positively selected more efficiently, thereby accelerating the onset of spontaneous autoimmune arthritis. Our findings highlight dual TCR-ß expression as a mechanism that can enhance the maturation of autoreactive pathogenic T cells and lead to more rapid development of autoimmune disease.


Asunto(s)
Alelos , Artritis/genética , Artritis/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Antígenos CD4/genética , Antígenos CD4/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocarditis/genética , Miocarditis/inmunología , Autotolerancia/genética , Autotolerancia/inmunología , Subgrupos de Linfocitos T/inmunología , Timocitos/inmunología , Transgenes
9.
J Immunol ; 187(5): 2702-10, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21795599

RESUMEN

The immunopathogenic mechanisms mediating inflammation in multiorgan autoimmune diseases may vary between the different target tissues. We used the K/BxN TCR transgenic mouse model to investigate the contribution of CD4(+) T cells and ß(2) integrins in the pathogenesis of autoimmune arthritis and endocarditis. Depletion of CD4(+) T cells following the onset of arthritis specifically prevented the development of cardiac valve inflammation. Genetic absence of ß(2) integrins had no effect on the severity of arthritis and unexpectedly increased the extent of cardiovascular pathology. The exaggerated cardiac phenotype of the ß(2) integrin-deficient K/BxN mice was accompanied by immune hyperactivation and was linked to a defect in regulatory T cells. These findings are consistent with a model in which the development of arthritis in K/BxN mice relies primarily on autoantibodies, whereas endocarditis depends on an additional contribution of effector T cells. Furthermore, strategies targeting ß(2) integrins for the treatment of systemic autoimmune conditions need to consider not only the role of these molecules in leukocyte recruitment to sites of inflammation, but also their impact on the regulation of immunological tolerance.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Antígenos CD18/inmunología , Miocarditis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocarditis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...