Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Immunol ; 195(3): 1202-17, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26109647

RESUMEN

Store-operated Ca(2+) entry (SOCE) through Ca(2+) release-activated Ca(2+) (CRAC) channels is essential for immunity to infection. CRAC channels are formed by ORAI1 proteins in the plasma membrane and activated by stromal interaction molecule (STIM)1 and STIM2 in the endoplasmic reticulum. Mutations in ORAI1 and STIM1 genes that abolish SOCE cause severe immunodeficiency with recurrent infections due to impaired T cell function. SOCE has also been observed in cells of the innate immune system such as macrophages and dendritic cells (DCs) and may provide Ca(2+) signals required for their function. The specific role of SOCE in macrophage and DC function, as well as its contribution to innate immunity, however, is not well defined. We found that nonselective inhibition of Ca(2+) signaling strongly impairs many effector functions of bone marrow-derived macrophages and bone marrow-derived DCs, including phagocytosis, inflammasome activation, and priming of T cells. Surprisingly, however, macrophages and DCs from mice with conditional deletion of Stim1 and Stim2 genes, and therefore complete inhibition of SOCE, showed no major functional defects. Their differentiation, FcR-dependent and -independent phagocytosis, phagolysosome fusion, cytokine production, NLRP3 inflammasome activation, and their ability to present Ags to activate T cells were preserved. Our findings demonstrate that STIM1, STIM2, and SOCE are dispensable for many critical effector functions of macrophages and DCs, which has important implications for CRAC channel inhibition as a therapeutic strategy to suppress pathogenic T cells while not interfering with myeloid cell functions required for innate immunity.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/metabolismo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Calcio/metabolismo , Canales de Calcio/genética , Proteínas de Unión al Calcio/inmunología , Proteínas Portadoras/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Retículo Endoplásmico/metabolismo , Humanos , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Activación de Linfocitos/inmunología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína ORAI1 , Fagocitosis/inmunología , Inmunodeficiencia Combinada Grave/genética , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2 , Linfocitos T/inmunología
3.
Cell Rep ; 24(11): 3045-3060.e5, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30208327

RESUMEN

T cell acute lymphoblastic leukemia (T-ALL) is commonly associated with activating mutations in the NOTCH1 pathway. Recent reports have shown a link between NOTCH1 signaling and intracellular Ca2+ homeostasis in T-ALL. Here, we investigate the role of store-operated Ca2+ entry (SOCE) mediated by the Ca2+ channel ORAI1 and its activators STIM1 and STIM2 in T-ALL. Deletion of STIM1 and STIM2 in leukemic cells abolishes SOCE and significantly prolongs the survival of mice in a NOTCH1-dependent model of T-ALL. The survival advantage is unrelated to the leukemic cell burden but is associated with the SOCE-dependent ability of malignant T lymphoblasts to cause inflammation in leukemia-infiltrated organs. Mice with STIM1/STIM2-deficient T-ALL show a markedly reduced necroinflammatory response in leukemia-infiltrated organs and downregulation of signaling pathways previously linked to cancer-induced inflammation. Our study shows that leukemic T lymphoblasts cause inflammation of leukemia-infiltrated organs that is dependent on SOCE.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Animales , Femenino , Inflamación/genética , Ratones , Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 2/genética
4.
Nat Commun ; 8: 14714, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294127

RESUMEN

Store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels is critical for lymphocyte function and immune responses. CRAC channels are hexamers of ORAI proteins that form the channel pore, but the contributions of individual ORAI homologues to CRAC channel function are not well understood. Here we show that deletion of Orai1 reduces, whereas deletion of Orai2 increases, SOCE in mouse T cells. These distinct effects are due to the ability of ORAI2 to form heteromeric channels with ORAI1 and to attenuate CRAC channel function. The combined deletion of Orai1 and Orai2 abolishes SOCE and strongly impairs T cell function. In vivo, Orai1/Orai2 double-deficient mice have impaired T cell-dependent antiviral immune responses, and are protected from T cell-mediated autoimmunity and alloimmunity in models of colitis and graft-versus-host disease. Our study demonstrates that ORAI1 and ORAI2 form heteromeric CRAC channels, in which ORAI2 fine-tunes the magnitude of SOCE to modulate immune responses.


Asunto(s)
Calcio/metabolismo , Inmunidad , Proteína ORAI2/metabolismo , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Proliferación Celular , Colitis/inmunología , Colitis/patología , Citocinas/biosíntesis , Eliminación de Gen , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Homeostasis , Humanos , Inmunidad Humoral , Activación del Canal Iónico , Recuento de Linfocitos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Proteína ORAI1/deficiencia , Proteína ORAI1/metabolismo , Proteína ORAI2/deficiencia , Multimerización de Proteína , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo
5.
Channels (Austin) ; 7(5): 379-91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23511024

RESUMEN

Store-operated Ca(2+) entry (SOCE) is an important Ca(2+) influx pathway, which is defined by the fact that depletion of intracellular Ca(2+) stores, mainly the endoplasmic reticulum (ER), triggers the opening of Ca(2+) channels in the plasma membrane. The best characterized SOC channel is the Ca(2+) release-activated Ca(2+) (CRAC) channel, which was first described in cells of the immune system but has since been reported in many different cell types. CRAC channels are multimers of ORAI family proteins, of which ORAI1 is the best characterized. They are activated by stromal interaction molecules (STIM) 1 and 2, which respond to the depletion of intracellular Ca(2+) stores with oligomerization and binding to ORAI proteins. The resulting SOCE is critical for the physiological function of many cell types including immune cells and platelets. Recent studies using cell lines, animal models and primary cells from human patients with defects in SOCE have highlighted the importance of this Ca(2+) entry mechanism in a variety of pathophysiological processes. This review focuses on the role of SOCE in immunity to infection, allergy, hemostasis and cancer.


Asunto(s)
Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Inmunidad Adaptativa , Plaquetas/fisiología , Hemostasis , Humanos , Inmunidad Innata , Infecciones/inmunología , Neoplasias/metabolismo , Proteína ORAI1 , Molécula de Interacción Estromal 1
6.
J Neurotrauma ; 30(22): 1919-24, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23819447

RESUMEN

Astrogliosis occurs following injury to the zebra finch brain. To date, only estrogen synthase (aromatase) has been identified in injury-induced astrocytes. The expression of other steroidogenic enzymes or their receptors remains unknown in the avian brain. However, in mammals, an upregulation of androgen receptors has been identified in glial cells. The aim of this study was to determine if the androgen receptor is upregulated following injury in adult zebra finches. Finches were given a single penetrating injury and brain tissue was collected 24 or 72 h later. Expression of androgen receptor was examined using immunohistochemistry and quantified using quantitative polymerase chain reaction (qPCR) analysis. Androgen receptors were localized to astrocytes versus neurons, further solidifying the role for astrocytes in neural recovery.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Pinzones/metabolismo , Receptores Androgénicos/biosíntesis , Regulación hacia Arriba , Animales , Astrocitos/patología , Encéfalo/citología , Encéfalo/patología , Lesiones Encefálicas/genética , Lesiones Encefálicas/fisiopatología , Femenino , Pinzones/lesiones , Masculino , Receptores Androgénicos/genética , Recuperación de la Función/genética , Factores Sexuales , Factores de Tiempo , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA