Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunity ; 34(4): 492-504, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21497118

RESUMEN

Polarized segregation of proteins in T cells is thought to play a role in diverse cellular functions including signal transduction, migration, and directed secretion of cytokines. Persistence of this polarization can result in asymmetric segregation of fate-determining proteins during cell division, which may enable a T cell to generate diverse progeny. Here, we provide evidence that a lineage-determining transcription factor, T-bet, underwent asymmetric organization in activated T cells preparing to divide and that it was unequally partitioned into the two daughter cells. This unequal acquisition of T-bet appeared to result from its asymmetric destruction during mitosis by virtue of concomitant asymmetric segregation of the proteasome. These results suggest a mechanism by which a cell may unequally localize cellular activities during division, thereby imparting disparity in the abundance of cell fate regulators in the daughter cells.


Asunto(s)
Mitosis , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Dominio T Box/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Polaridad Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas de Dominio T Box/metabolismo , Linfocitos T/enzimología
2.
Immunity ; 30(3): 372-83, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19285436

RESUMEN

Intracellular cyclic adenosine monophosphate (cAMP) suppresses innate immunity by inhibiting proinflammatory cytokine production from monocytic cells. Enhanced expression of interleukin-10 (IL-10) has been suggested to be the mechanism of suppression. However, cAMP is still capable of suppressing production of the cytokines TNF-alpha and IL-12 in IL-10-deficient dendritic cells (DCs). Here, we demonstrated that the transcription factor c-Fos was responsible for the cAMP-mediated suppression of inflammatory cytokine production. c-Fos accumulated at high amounts in response to cAMP and lipopolysaccharide (LPS). Overexpression of c-Fos suppressed LPS-induced cytokine production, whereas cAMP-mediated suppression of TNF-alpha and IL-12 was impaired in Fos(-/-) DCs or in RAW264.7 cells treated with c-Fos siRNA. c-Fos physically interacted with p65 protein and reduced the recruitment of p65 to the Tnf promoter. Multiple sites of c-Fos were phosphorylated by the IKKbeta protein. Thus, we propose that c-Fos is a substrate of IKKbeta and is responsible for the immunosuppressive effect of cAMP.


Asunto(s)
AMP Cíclico/inmunología , Citocinas/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-fos/inmunología , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-fos/clasificación , Proteínas Proto-Oncogénicas c-fos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Immunity ; 29(6): 971-85, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-19100702

RESUMEN

Although T lymphocytes are constitutively nonadherent cells, they undergo facultative polarity during migration and upon interaction with cells presenting cognate antigen, suggesting that cell polarity might be critical for target cell destruction. Using two-photon imaging of tumor-infiltrating T lymphocytes, we found that CD44, a receptor for extracellular matrix proteins and glycosaminoglycans, was crucial for interstitial T cell navigation and, consequently, efficient tumor cell screening. CD44 functioned as a critical regulator of intratumoral movement by stabilizing cell polarity in migrating T cells, but not during target cell interactions. Stable anterior-posterior asymmetry was maintained by CD44 independently of its extracellular domain. Instead, migratory polarity depended on the recruitment of ezrin, radixin, moesin (ERM) proteins by the intracellular domain of CD44 to the posterior cellular protrusion. Our results formally demonstrate that CD44-dependent T lymphocyte locomotion within target sites represents an essential immunologic checkpoint that determines the potency of T cell effector functions.


Asunto(s)
Movimiento Celular/inmunología , Polaridad Celular/inmunología , Receptores de Hialuranos/inmunología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Adhesión Celular/inmunología , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Movimiento Celular/genética , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/inmunología , Proteínas de Microfilamentos/metabolismo , Neoplasias/metabolismo , Estructura Terciaria de Proteína , Linfocitos T Citotóxicos/metabolismo
4.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28449314

RESUMEN

Moxetumomab pasudotox is a second-generation recombinant immunotoxin against CD22 on B-cell lineages. Antileukemic activity has been demonstrated in children with chemotherapy-refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL), with variable responses. Here, we report in vitro and in vivo evaluation of moxetumomab pasudotox treatment of human cell lines and patient-derived cells as a preliminary study to understand characteristics of sensitivity to treatment. Binding, internalization, and apoptosis were evaluated using fluorescently tagged moxetumomab pasudotox. Studies in NOD-scid IL2Rgnull mice showed a modest survival benefit in mice engrafted with 697 cells but not in NALM6 or the two patient-derived xenograft models.


Asunto(s)
Apoptosis/efectos de los fármacos , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Adolescente , Adulto , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
5.
J Exp Med ; 203(4): 1021-31, 2006 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-16606674

RESUMEN

Suppressor of cytokine signaling (SOCS)3 is a major negative feedback regulator of signal transducer and activator of transcription (STAT)3-activating cytokines. Transgenic mouse studies indicate that high levels of SOCS3 in T cells result in type 2 T helper cell (Th2) skewing and lead to hypersensitivity to allergic diseases. To define the physiological roles of SOCS3 in T cells, we generated T cell-specific SOCS3 conditional knockout mice. We found that the mice lacking SOCS3 in T cells showed reduced immune responses not only to ovalbumin-induced airway hyperresponsiveness but also to Leishmania major infection. In vitro, SOCS3-deficient CD4+ T cells produced more transforming growth factor (TGF)-beta1 and interleukin (IL)-10, but less IL-4 than control T cells, suggesting preferential Th3-like differentiation. We found that STAT3 positively regulates TGF-beta1 promoter activity depending on the potential STAT3 binding sites. Furthermore, chromatin immunoprecipitation assay revealed that more STAT3 was recruited to the TGF-beta1 promoter in SOCS3-deficient T cells than in control T cells. The activated STAT3 enhanced TGF-beta1 and IL-10 expression in T cells, whereas the dominant-negative form of STAT3 suppressed these. From these findings, we propose that SOCS3 regulates the production of the immunoregulatory cytokines TGF-beta1 and IL-10 through modulating STAT3 activation.


Asunto(s)
Regulación hacia Abajo/inmunología , Interleucina-10/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética , Linfocitos T Colaboradores-Inductores/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Diferenciación Celular/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/fisiología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1 , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
6.
J Immunol ; 185(12): 7151-5, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21076068

RESUMEN

A hallmark of autoimmune lymphoproliferative syndrome (ALPS), caused by mutation of the Fas death receptor, is massive lymphadenopathy from aberrant expansion of CD4(-)CD8(-) (double-negative [DN]) T cells. Eomesodermin (Eomes) is a member of the T-box family of transcription factors and plays critical roles in effector cell function and memory cell fitness of CD8(+) T lymphocytes. We provide evidence in this study that DN T cells exhibit dysregulated expression of Eomes in humans and mice with ALPS. We also find that T cell-specific deletion of Eomes prevents lymphoid hypertrophy and accumulation of DN T cells in Fas-mutant mice. Although Eomes has critical physiological roles in the function and homeostasis of CD8(+) T cells, overexpression of Eomes appears to enable pathological induction or expansion of unusual CD8-related T cell subsets. Thus, antagonism of Eomes emerges as a therapeutic target for DN T cell ablation in ALPS.


Asunto(s)
Síndrome Linfoproliferativo Autoinmune/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Dominio T Box/inmunología , Subgrupos de Linfocitos T/inmunología , Receptor fas , Animales , Síndrome Linfoproliferativo Autoinmune/genética , Síndrome Linfoproliferativo Autoinmune/patología , Síndrome Linfoproliferativo Autoinmune/terapia , Linfocitos T CD8-positivos/patología , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Masculino , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/genética , Subgrupos de Linfocitos T/patología
7.
Immunol Rev ; 221: 147-62, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18275480

RESUMEN

Recent advances in two-photon microscopy have provided a new way of visualizing the behavior of fluorescently tagged cells within their natural microenvironment. This technology has allowed for generating a detailed picture of the cellular interaction dynamics operant in the activation of T cells and B cells during primary immune responses within secondary lymphoid organs. In contrast, relatively little is known about the migratory and interactive behavior of effector T cells within peripheral organs. We have recently developed a two-photon microscopy model that enables tracking of cytotoxic T cells within tumors. We have demonstrated that tumor-infiltrating T lymphocytes (TILs) follow random migratory paths and that their migratory properties depend on signals from the T-cell receptor. We further showed that TILs underwent short- and long-term interactions with tumor cells as well as macrophages. Recently, we succeeded in dynamic imaging of the distribution of fluorescently tagged molecules within TILs at subcellular resolution, which will be instrumental for defining the composition of the lytic synapse as well as the targeted release of cytotoxic granules by these cells. The purpose of this review is to put our findings into the context of the current literature and to point out the molecular cues mediating effector T-cell function as candidates for future investigation.


Asunto(s)
Linfocitos Infiltrantes de Tumor/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neoplasias Experimentales/patología , Linfocitos T/patología , Animales , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología
8.
Cancer Sci ; 100(4): 730-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19469017

RESUMEN

Inflammation has been shown to contribute to both tumor development and antitumor immunity. However, conditions determining these opposing effects are not well understood. Suppressor of cytokine signaling 1 (SOCS1) has been shown to play an important role in regulating inflammation and tumor development. It has been reported that silencing of SOCS1 gene in dendritic cells potentiates antitumor immunity, while SOCS1-deficiency in whole organs except for T and B cells enhances inflammation-mediated colon tumor development. To determine which types of cells are important for the suppression of tumor development by SOCS1-deficiency, we employed the conditional knockout strategy. SOCS1 gene was deleted in macrophages and neutrophils by crossing SOCS1-flox/flox mice with LysM-cre mice. Resulting conditional knockout (cKO) mice showed enhanced sensitivity to endotoxin shock. SOCS1-cKO mice survived much longer than wild-type mice after B16 melanoma transplantation. Colon carcinogenesis induced by 1,2-dimethylhydrazine (DMH) plus dextran sulfate sodium (DSS) was also reduced in SOCS1-cKO mice. SOCS1-deficiency in monocytic cells enhanced tumor-killing activity of macrophages and tumor-specific cytotoxic T cell activity. These results suggest that inflammation induced by SOCS1-deficiency in monocytes potentiates antitumor immune responses rather than tumor-promoting inflammation.


Asunto(s)
Metilación de ADN , Inflamación/inmunología , Macrófagos/inmunología , Melanoma Experimental/inmunología , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Inflamación/genética , Estimación de Kaplan-Meier , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Leukoc Biol ; 105(4): 741-753, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30702754

RESUMEN

Infiltration of acute lymphoblastic leukemia (ALL) blasts into the CNS remains as a major clinical problem, with high risk for chemotherapy-resistant relapse and treatment-related morbidity. Despite the common inclusion of CNS prophylaxis treatments in therapy regimens, there are significant gaps in understanding the mechanisms that mediate leukemia cell entry into the CNS as well as roles for resident cells in the brain. In this study, we employ a xenograft model of human B cell precursor (BCP)-ALL in immunocompromised mice. This model system recapitulates key pathological characteristics of leptomeningeal involvement seen in patients and provides insights into rare cases that involve parenchymal invasion. We examine the infiltration of engrafted leukemia blasts into brains of recipient mice and provide evidence that the interaction between blasts and brain resident cells causes aberrant activation of host cells in the brain microenvironment. BCP-ALL blasts also release multiple cytokines and exosomes containing IL-15 that bind and are internalized by astrocytes and brain vessel endothelial cells. Leukemic invasion is linked to production of VEGF-AA by astrocytes and disruption of the blood-brain-barrier (BBB) integrity. Knockdown of either IL-15 or IL-15Rα in the NALM6 cell line decreases CNS infiltration in engrafted mice. These results provide important insights into the multiple mechanisms by which lymphoblasts modulate the brain microenvironment to breach the BBB for metastatic invasion.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Citocinas/metabolismo , Exosomas/metabolismo , Leucemia/patología , Animales , Barrera Hematoencefálica/patología , Línea Celular Tumoral , Citocinas/sangre , Exosomas/ultraestructura , Humanos , Interleucina-15/metabolismo , Leucemia/sangre , Ratones , Metástasis de la Neoplasia , Transducción de Señal , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Signal ; 9(456): ra116, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27899526

RESUMEN

The pre-B cell receptor (pre-BCR) is an immature form of the BCR critical for early B lymphocyte development. It is composed of the membrane-bound immunoglobulin (Ig) heavy chain, surrogate light chain components, and the signaling subunits Igα and Igß. We developed monovalent quantum dot (QD)-labeled probes specific for Igß to study the behavior of pre-BCRs engaged in autonomous, ligand-independent signaling in live B cells. Single-particle tracking revealed that QD-labeled pre-BCRs engaged in transient, but frequent, homotypic interactions. Receptor motion was correlated at short separation distances, consistent with the formation of dimers and higher-order oligomers. Repeated encounters between diffusing pre-BCRs appeared to reflect transient co-confinement in plasma membrane domains. In human B cell precursor acute lymphoblastic leukemia (BCP-ALL) cells, we showed that frequent, short-lived, homotypic pre-BCR interactions stimulated survival signals, including expression of BCL6, which encodes a transcriptional repressor. These survival signals were blocked by inhibitory monovalent antigen-binding antibody fragments (Fabs) specific for the surrogate light chain components of the pre-BCR or by inhibitors of the tyrosine kinases Lyn and Syk. For comparison, we evaluated pre-BCR aggregation mediated by dimeric galectin-1, which has binding sites for carbohydrate and for the surrogate light chain λ5 component. Galectin-1 binding resulted in the formation of large, highly immobile pre-BCR aggregates, which was partially relieved by the addition of lactose to prevent the cross-linking of galectin-BCR complexes to other glycosylated membrane components. Analysis of the pre-BCR and its signaling partners suggested that they could be potential targets for combination therapy in BCP-ALL.


Asunto(s)
Receptores de Células Precursoras de Linfocitos B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Precursoras de Linfocitos B/inmunología , Multimerización de Proteína/inmunología , Transducción de Señal/inmunología , Línea Celular Tumoral , Supervivencia Celular/inmunología , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Precursoras de Linfocitos B/patología , Proteínas Proto-Oncogénicas c-bcl-6/inmunología , Quinasa Syk/inmunología , Familia-src Quinasas/inmunología
11.
Nat Commun ; 6: 6301, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25709008

RESUMEN

The precise pathways of memory T-cell differentiation are incompletely understood. Here we exploit transgenic mice expressing fluorescent cell cycle indicators to longitudinally track the division dynamics of individual CD8(+) T cells. During influenza virus infection in vivo, naive T cells enter a CD62L(intermediate) state of fast proliferation, which continues for at least nine generations. At the peak of the anti-viral immune response, a subpopulation of these cells markedly reduces their cycling speed and acquires a CD62L(hi) central memory cell phenotype. Construction of T-cell family division trees in vitro reveals two patterns of proliferation dynamics. While cells initially divide rapidly with moderate stochastic variations of cycling times after each generation, a slow-cycling subpopulation displaying a CD62L(hi) memory phenotype appears after eight divisions. Phenotype and cell cycle duration are inherited by the progeny of slow cyclers. We propose that memory precursors cell-intrinsically modulate their proliferative activity to diversify differentiation pathways.


Asunto(s)
Linfocitos T CD8-positivos/citología , Ciclo Celular , Diferenciación Celular , Animales , Linfocitos T CD8-positivos/fisiología , Perfilación de la Expresión Génica , Genes Reporteros , Ratones Endogámicos C57BL , Ratones Transgénicos , Transcriptoma
12.
Pigment Cell Melanoma Res ; 27(5): 764-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24902993

RESUMEN

Solid cancers are composed of heterogeneous zones containing proliferating and quiescent cells. Despite considerable insight into the molecular mechanisms underlying aberrant cell cycle progression, there is limited understanding of the relationship between the cell cycle on the one side, and melanoma cell motility, invasion, and drug sensitivity on the other side. Utilizing the fluorescent ubiquitination-based cell cycle indicator (FUCCI) to longitudinally monitor proliferation and migration of melanoma cells in 3D culture and in vivo, we found that invading melanoma cells cycle actively, while G1-arrested cells showed decreased invasion. Melanoma cells in a hypoxic environment or treated with mitogen-activated protein kinase pathway inhibitors remained G1-arrested for extended periods of time, with proliferation and invasion resuming after re-exposure to a more favorable environment. We challenge the idea that the invasive and proliferative capacity of melanoma cells are mutually exclusive and further demonstrate that a reversibly G1-arrested subpopulation survives in the presence of targeted therapies.


Asunto(s)
Melanoma/patología , Invasividad Neoplásica , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Colorantes Fluorescentes/química , Fase G1 , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Ubiquitina/química
15.
Science ; 315(5819): 1687-91, 2007 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-17332376

RESUMEN

A hallmark of mammalian immunity is the heterogeneity of cell fate that exists among pathogen-experienced lymphocytes. We show that a dividing T lymphocyte initially responding to a microbe exhibits unequal partitioning of proteins that mediate signaling, cell fate specification, and asymmetric cell division. Asymmetric segregation of determinants appears to be coordinated by prolonged interaction between the T cell and its antigen-presenting cell before division. Additionally, the first two daughter T cells displayed phenotypic and functional indicators of being differentially fated toward effector and memory lineages. These results suggest a mechanism by which a single lymphocyte can apportion diverse cell fates necessary for adaptive immunity.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , División Celular , Memoria Inmunológica , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Antígenos CD/análisis , Antígenos CD8/análisis , Diferenciación Celular , Linaje de la Célula , Polaridad Celular , Células Dendríticas/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitosis , Proteínas del Tejido Nervioso/análisis , Proteína Quinasa C/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Interferón/análisis , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Receptor de Interferón gamma
16.
J Immunol ; 177(8): 5377-85, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015723

RESUMEN

Recent lines of evidence have demonstrated that IL-27, a newly identified IL-12-related cytokine, has two apparently conflicting roles in immune responses: one as an initiator of Th1 responses and the other as an attenuator of inflammatory cytokine production. Although the IL-27-mediated Th1 initiation mechanism has been elucidated, little is known about the molecular basis for the suppression of cytokine production. In the present study, we demonstrated that IL-27 suppressed the production of various proinflammatory cytokines by fully activated CD4+ T cells while it had no effect on the cytokine production by CD4+ T cells at early phases of activation. IL-27 also suppressed IL-17 production by activated CD4+ T cells, thereby counteracting IL-23, another IL-12-related cytokine with proinflammatory effects. In fully activated CD4+ T cells, STAT3 was preferentially activated by IL-27 stimulation, whereas both STAT1 and 3 were activated by IL-27 in early activated CD4+ T cells. Lack of STAT3 in fully activated cells impaired the suppressive effects of IL-27. These data indicated that the preferential activation of STAT3 in fully activated CD4+ T cells plays an important role in the cytokine suppression by IL-27/WSX-1.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Interleucina-17/biosíntesis , Interleucina-23/fisiología , Interleucinas/fisiología , Factor de Transcripción STAT3/fisiología , Células TH1/citología , Animales , Línea Celular , Citocinas/biosíntesis , Humanos , Activación de Linfocitos , Ratones , Ratones Noqueados , Receptores de Citocinas/fisiología , Receptores de Interleucina , Factor de Transcripción STAT3/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 290(5): L971-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16373672

RESUMEN

Neuromedin U (NMU) is a neuropeptide expressed not only in the central nervous system but also in various organs, including the gastrointestinal tract and lungs. NMU interacts with two G protein-coupled receptors, NMU-R1 and NMU-R2. Although NMU-R2 is expressed in a specific region of the brain, NMU-R1 is expressed in various peripheral tissues, including immune and hematopoietic cells. Our recent study demonstrated an important role of NMU in mast cell-mediated inflammation. In this study, we showed that airway eosinophilia was reduced in NMU-deficient mice in an allergen-induced asthma model. There were no differences in the antigen-induced Th2 responses between wild-type and NMU knockout mice. NMU-R1 was highly expressed in the eosinophil cell line, and NMU directly induced Ca(2+) mobilization and extracellular/signal-regulated kinase phosphorylation. NMU also induced cell adhesion to components of the extracellular matrix (fibronectin and collagen type I), and chemotaxis in vitro. Furthermore, NMU-R1 was also expressed in human peripheral blood eosinophils, and NMU induced cell adhesion in a dose-dependent manner. These data indicate that NMU promotes eosinophil infiltration into inflammatory sites by directly activating eosinophils. Our study suggests that NMU receptor antagonists could be novel targets for pharmacological inhibition of allergic inflammatory diseases, including asthma.


Asunto(s)
Alérgenos , Eosinofilia/fisiopatología , Eosinófilos/fisiología , Neuropéptidos/fisiología , Animales , Líquido del Lavado Bronquioalveolar/citología , Señalización del Calcio/fisiología , Adhesión Celular/inmunología , Adhesión Celular/fisiología , Eosinofilia/inmunología , Eosinofilia/patología , Eosinófilos/inmunología , Eosinófilos/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Macrófagos Alveolares/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia , Neuropéptidos/genética , Receptores de Neurotransmisores/antagonistas & inhibidores , Receptores de Neurotransmisores/efectos de los fármacos
18.
J Immunol ; 168(12): 6404-11, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12055259

RESUMEN

Previous studies have shown that IL-10 can induce the expression of the suppressor of cytokine signaling 3 (SOCS-3) mRNA in human monocytes and neutrophils, suggesting that the capacity of IL-10 to inhibit the expression of LPS-inducible proinflammatory genes may depend on SOCS-3 induction. However, no direct experimental evidence has been provided to support such hypothesis. Herein, we show that stable transfection of SOCS-3 into the mouse macrophage cell line J774 resulted in an inhibition of NO, TNF-alpha, IL-6, and GM-CSF secretion in response to LPS at levels similar to those exerted by IL-10 in LPS-stimulated wild-type J774. Constitutive SOCS-3 expression also down-regulated the mRNA expression of inducible NO synthase and IL-6 and impaired the production of TNF-alpha, mainly at a post-transcriptional level. In addition, SOCS-3-transfected cells displayed a constitutive expression of the IL-1R antagonist gene, consistent with the observation that IL-10 enhances IL-1R antagonist mRNA in LPS-stimulated wild-type cells. Furthermore, in peritoneal macrophages harvested from mice carrying heterozygous disruption of the SOCS-3 gene, IL-10 was less effective in repressing LPS-stimulated TNF-alpha and NO production. Taken together, our data show that SOCS-3 inhibits LPS-induced macrophage activation, strongly supporting the idea that it plays a role in the molecular mechanism by which IL-10 down-modulates the effector functions of LPS-activated macrophages. Finally, we show that forced expression of SOCS-3 significantly suppresses the ability of IL-10 to trigger tyrosine phosphorylation of STAT3. Therefore, SOCS-3 functions both as an LPS signal inhibitor and as a negative feedback regulator of IL-10/STAT3 signaling.


Asunto(s)
Interleucina-10/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Activación de Macrófagos/inmunología , Proteínas/fisiología , Proteínas Represoras , Transducción de Señal/inmunología , Factores de Transcripción , Animales , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-10/fisiología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Activación de Macrófagos/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Factor de Transcripción STAT3 , Sialoglicoproteínas/biosíntesis , Sialoglicoproteínas/genética , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
19.
J Biol Chem ; 279(14): 13746-54, 2004 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-14742442

RESUMEN

SOCS-3 (suppressor of cytokine signaling 3) is an intracellular protein that is selectively and rapidly induced by appropriate agonists and that modulates responses of immune cells to cytokines by interfering with the Janus kinase/signal transducer and activator of transcription (Jak/STAT) pathway. On the basis of the observations that interferon gamma (IFNgamma) up-regulates SOCS-3 gene and protein expression in primary mouse macrophages, J774 macrophage cell line and embryonal fibroblasts, we investigated which sequences of the 5' SOCS-3 gene are responsive to IFNgamma. By promoter deletion analysis we identified a functional IFNgamma-responsive element, located at nucleotides -72/-64 upstream from the transcription initiation, whose presence and integrity is necessary to ensure responsiveness to IFNgamma. This element contains a STAT consensus binding sequence (SOCS-3/STAT-binding element (SBE)) whose specific mutation totally abolished the responsiveness to IFNgamma. In contrast, discrete deletion of other 5' regions of the SOCS-3 promoter did not substantially modify the inducibility by IFNgamma. Electromobility shift assay analyses revealed that IFNgamma promotes specific DNA binding activities to an oligonucleotide probe containing the SOCS-3/SBE sequence. Even though IFNgamma triggered tyrosine phosphorylation of both STAT1 and STAT3 in macrophages and J774 cells, only STAT1 was appropriately activated and thus found to specifically bind to the SOCS-3/SBE oligonucleotide probe. Accordingly, IFNgamma-induced SOCS-3 protein expression was not impaired in STAT3-deficient embryonal fibroblasts. Taken together, these results demonstrate that the induction of SOCS-3 by IFNgamma depends upon the presence of a STAT-binding element in the SOCS-3 promoter that is specifically activated by STAT1.


Asunto(s)
Antineoplásicos/farmacología , Interferón gamma/farmacología , Macrófagos Peritoneales/fisiología , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Macrófagos Peritoneales/citología , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/metabolismo
20.
J Biol Chem ; 279(8): 6905-10, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14699146

RESUMEN

The suppressor of cytokine signaling-3 (SOCS3/CIS3) has been shown to be an important negative regulator of cytokines, especially cytokines that activate STAT3. To examine the role of SOCS3 in neutrophils and the granulocyte colony-stimulating factor (G-CSF) signaling in vivo, we compared neutrophils from two types of conditional knockout mice, LysM-Cre:SOCS3(fl/fl) mice and Tie2-Cre:SOCS3(fl/fl) mice, in which the Socs3 gene had been deleted in mature neutrophils and hematopoietic stem cells, respectively. The size of the G-CSF-dependent colonies from Tie2-Cre:SOCS3(fl/fl) mouse bone marrow was much larger than that of colonies from control wild-type mice, while the size of interleukin-3-dependent colonies was similar. Moreover, LysM-Cre:SOCS3(fl/fl) mice had more neutrophils than SOCS3(fl/fl) mice, suggesting that SOCS3 is a negative regulator of G-CSF signaling in neutrophils. Consistent with this notion, G-CSF-induced STAT3 as well as mitogen-activated protein kinase activation was much stronger and prolonged in SOCS3-deficient mature neutrophils than in wild-type neutrophils. The preventive effect of G-CSF on apoptosis was more prominent in SOCS3-deficient mature neutrophils than in control neutrophils. These data indicate that SOCS3 negatively regulates granulopoiesis and G-CSF signaling in neutrophils and may contribute to neutrophilia or neutropenia.


Asunto(s)
Granulocitos/fisiología , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Animales , Apoptosis , Western Blotting , Quimiotaxis , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Exones , Citometría de Flujo , Eliminación de Gen , Interleucina-3/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Genéticos , Neutrófilos/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3 , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Factores de Tiempo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA