Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(41): 11543-11548, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27671645

RESUMEN

Adenine-thymine (AT)-rich interactive domain containing protein 5a (Arid5a) is an RNA-binding protein that has been shown to play an important immune regulatory function via the stabilization of IL-6 and STAT3 mRNA. However, the role of Arid5a in the overwhelming and uncontrolled immune response that leads to septic shock is unknown. Here, we report that Arid5a-deficient mice are highly resistant to lipopolysaccharide (LPS)-induced endotoxic shock and secrete lower levels of major proinflammatory cytokines, including IFN-γ, IL-6, and TNF-α, than WT mice in response to LPS. Arid5a deficiency resulted in decreased levels of IFN-γ under Th1 cell conditions, in which T-box expressed in T cells (T-bet) mRNA expression was inhibited. Arid5a bound to the conserved stem loop structure of the 3'UTR of T-bet and stabilized its mRNA. Arid5a-deficient mice were also resistant to Propionibacterium acnes-primed LPS injection, which is considered to be a T-cell-mediated IFN-γ dependent endotoxic shock mouse model. Thus, regulation of IFN-γ by Arid5a via the stabilization of T-bet mRNA in Th1 cells contributes to the development of septic shock in mice. In addition, our previous study suggests that Arid5a control the IL-6 level in vivo in response to LPS by stabilization of IL-6 mRNA. We also observed that neutralization of IFN-γ and IL-6 significantly recovered the mice from endotoxic shock. Taken together, we conclude that Arid5a regulates the augmentation of IL-6 and IFN-γ in response to LPS, which possibly works synergistically for amplification of various other cytokines that ultimately cause the development of septic shock in mice.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Interferón gamma/metabolismo , Estabilidad del ARN/genética , Choque Séptico/metabolismo , Proteínas de Dominio T Box/genética , Factores de Transcripción/metabolismo , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Separación Celular , Secuencia Conservada/genética , Citocinas/sangre , Proteínas de Unión al ADN/deficiencia , Femenino , Células HEK293 , Humanos , Lipopolisacáridos , Activación de Linfocitos , Ratones Endogámicos C57BL , Pruebas de Neutralización , Conformación de Ácido Nucleico , Propionibacterium acnes/fisiología , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Choque Séptico/sangre , Choque Séptico/inmunología , Choque Séptico/microbiología , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Factores de Transcripción/deficiencia
2.
Int Immunol ; 29(2): 79-85, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28379390

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are among the major causes of death worldwide due to acute inflammation in the lung. AT-rich interactive domain-containing 5a (Arid5a) is an RNA-binding protein involved in inflammatory autoimmune disease through post-transcriptional control of Il6, Stat3 and Tbx21 gene expression. We found that Arid5a-deficient mice were highly refractory to bleomycin (BLM)-induced lethality. Arid5a deficiency suppressed lung pathology, cytokine production (especially, IL-6), and clinical symptoms in BLM-treated mice. Production of reactive oxygen species (ROS) in response to BLM-induced cellular damage was inhibited in Arid5a-deficient mice, potentially affecting the level of oxidized 1-palmitoyl-2-arachidonoyl-phosphaticylcholine (OxPAPC) production. OxPAPC, which is supposed to be a TLR4/TLR2 ligand, stimulated expression of the Arid5a and Il6 genes. Thus, reduction of ROS production in Arid5a-deficient mice could mitigate OxPAPC production, which in turn decreases IL-6 production in vivo due to dysregulated post-transcriptional regulation by loss of Arid5a. Therefore, the control of Arid5a expression represents a potential therapeutic target for treatment of ALI and ARDS.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Proteínas de Unión al ADN/genética , Pulmón/patología , Neumonía/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Factores de Transcripción/genética , Lesión Pulmonar Aguda/inducido químicamente , Animales , Bleomicina/administración & dosificación , Humanos , Interleucina-6/metabolismo , Pulmón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/terapia , Especies Reactivas de Oxígeno/metabolismo , Síndrome de Dificultad Respiratoria/terapia
3.
Int Immunol ; 28(6): 307-15, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26865412

RESUMEN

Thalidomide and its derivatives, collectively referred to as immunomodulatory drugs (IMiDs), are effective inhibitors of inflammation and are known to inhibit TLR-induced TNFα production. The identification of Cereblon as the receptor for these compounds has led to a rapid advancement in our understanding of IMiD properties; however, there remain no studies addressing the role of Cereblon in mediating the suppressive effect of IMiDs on TLR responses. Here, we developed Cereblon-deficient mice using the CRISPR-Cas9 system. TLR-induced cytokine responses were unaffected by Cereblon deficiency in vivo Moreover, IMiD treatment inhibited cytokine production even in the absence of Cereblon. The IMiD-induced suppression of cytokine production therefore occurs independently of Cereblon in mice. Further investigation revealed that IMiDs are potent inhibitors of TLR-induced type-1 interferon production via suppression of the TRIF/IRF3 pathway. These data suggest that IMiDs may prove effective in the treatment of disorders characterized by the ectopic production of type-1 interferon. Significantly, these properties are mediated separately from thalidomide's teratogenic receptor, Cereblon. Thus, certain therapeutic properties of Thalidomide can be separated from its harmful side effects.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Terapia de Inmunosupresión , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Lenalidomida , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Int Immunol ; 27(8): 405-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25862525

RESUMEN

Aryl hydrocarbon receptor (Ahr), a transcription factor, plays a critical role in autoimmune inflammation of the intestine. In addition, microRNAs (miRNAs), small non-coding oligonucleotides, mediate pathogenesis of inflammatory bowel diseases (IBD). However, the precise mechanism and interactions of these molecules in IBD pathogenesis have not yet been investigated. We analyzed the role of Ahr and Ahr-regulated miRNAs in colonic inflammation. Our results show that deficiency of Ahr in intestinal epithelial cells in mice exacerbated inflammation in dextran sodium sulfate-induced colitis. Deletion of Ahr in T cells attenuated colitis, which was manifested by suppressed Th17 cell infiltration into the lamina propria. Candidate miRNA analysis showed that induction of colitis elevated expression of the miR-212/132 cluster in the colon of wild-type mice, whereas in Ahr (-/-) mice, expression was clearly lower. Furthermore, miR-212/132(-/-) mice were highly resistant to colitis and had reduced levels of Th17 cells and elevated levels of IL-10-producing CD4(+) cells. In vitro analyses revealed that induction of type 1 regulatory T (Tr1) cells was significantly elevated in miR-212/132(-/-) T cells with increased c-Maf expression. Our findings emphasize the vital role of Ahr in intestinal homeostasis and suggest that inhibition of miR-212/132 represents a viable therapeutic strategy for treating colitis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Colitis/genética , Interleucina-10/genética , MicroARNs/genética , Receptores de Hidrocarburo de Aril/genética , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Proliferación Celular , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Sulfato de Dextran , Femenino , Regulación de la Expresión Génica , Homeostasis/inmunología , Interleucina-10/inmunología , Intestinos/inmunología , Intestinos/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/inmunología , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/inmunología , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/inmunología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células Th17/inmunología , Células Th17/patología
5.
Proc Natl Acad Sci U S A ; 110(23): 9409-14, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23676272

RESUMEN

Posttranscriptional regulation of IL-6 has been largely uncharacterized, with the exception of the ribonuclease Regnase-1, which prevents autoimmunity by destabilizing IL-6 mRNA. Here, we identified AT-rich interactive domain-containing protein 5A (Arid5a) as a unique RNA binding protein, which stabilizes IL-6 but not TNF-α mRNA through binding to the 3' untranslated region of IL-6 mRNA. Arid5a was enhanced in macrophages in response to LPS, IL-1ß, and IL-6. Arid5a deficiency inhibited elevation of IL-6 serum level in LPS-treated mice and suppressed IL-6 levels and the development of T(H)17 cells in experimental autoimmune encephalomyelitis. Importantly, Arid5a inhibited the destabilizing effect of Regnase-1 on IL-6 mRNA. These results indicate that Arid5a plays an important role in promotion of inflammatory processes and autoimmune diseases.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-6/inmunología , Estabilidad del ARN/inmunología , Proteínas de Unión al ARN/inmunología , Factores de Transcripción/inmunología , Regiones no Traducidas 3'/genética , Animales , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/metabolismo , Ensayo de Inmunoadsorción Enzimática , Interleucina-6/sangre , Lipopolisacáridos , Luciferasas , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasas/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(29): 11964-9, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23818645

RESUMEN

Aryl hydrocarbon receptor (AHR) plays critical roles in various autoimmune diseases such as multiple sclerosis by controlling interleukin-17 (IL-17)-producing T-helper (TH17) and regulatory T cells. Although various transcription factors and cytokines have been identified as key participants in TH17 generation, the role of microRNAs in this process is poorly understood. In this study, we found that expression of the microRNA (miR)-132/212 cluster is up-regulated by AHR activation under TH17-inducing, but not regulatory T-inducing conditions. Deficiency of the miR-132/212 cluster prevented the enhancement of TH17 differentiation by AHR activation. We also identified B-cell lymphoma 6, a negative regulator of TH17 differentiation, as a potential target of the miR-212. Finally, we investigated the roles of the miR-132/212 cluster in experimental autoimmune encephalomyelitis, a murine model of multiple sclerosis. Mice deficient in the miR-132/212 cluster exhibited significantly higher resistance to the development of experimental autoimmune encephalomyelitis and lower frequencies of both TH1 and TH17 cells in draining lymph nodes. Our findings reveal a unique mechanism of AHR-dependent TH17 differentiation that depends on the miR-132/212 cluster.


Asunto(s)
Diferenciación Celular/inmunología , Interleucina-17/metabolismo , MicroARNs/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Western Blotting , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Interleucina-17/inmunología , Luciferasas , Ratones , Ratones Noqueados , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Colaboradores-Inductores/metabolismo
7.
Int J Cancer ; 132(2): 459-71, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22532243

RESUMEN

Malignant pleural mesothelioma (MPM) is an aggressive tumor with poor prognosis for which an effective therapy remains to be established. This study investigated the therapeutic potential of gene delivery using suppressor of cytokine signaling 1 (SOCS-1), an endogenous inhibitor of intracellular signaling pathways, for the treatment of MPM. We infected MPM cells (MESO-4, H28 and H226) with adenovirus-expressing SOCS-1 vector to examine the effect of SOCS-1 overexpression on MPM cells. We evaluated the antitumor effect of SOCS-1 gene delivery combined with cisplatin plus pemetrexed by cell proliferation, apoptosis and invasion assay. We also investigated the regulation of NF-κB and STAT3 signaling related to apoptotic pathways. Furthermore, we evaluated the inhibition of tumor growth by SOCS-1 gene delivery combined with cisplatin plus pemetrexed in vivo. SOCS-1 gene delivery cooperated with cisplatin plus pemetrexed to inhibit cell proliferation, invasiveness and induction of apoptosis in MPM cells. SOCS-1 regulated NF-κB and STAT3 signaling to induce apoptosis in MESO-4 and H226 cells. Furthermore, SOCS-1 gene delivery cooperated with cisplatin plus pemetrexed to regulate NF-κB signaling and significantly inhibit tumor growth of MPM in vivo. These results suggest that SOCS-1 gene delivery has a potent antitumor effect against MPM and a potential for clinical use in combination with cisplatin plus pemetrexed.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mesotelioma/terapia , Neoplasias Pleurales/terapia , Proteínas Supresoras de la Señalización de Citocinas/genética , Adenoviridae/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Terapia Combinada , Activación Enzimática , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/análogos & derivados , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interferón gamma/farmacología , Interferón gamma/fisiología , Mesotelioma/patología , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , FN-kappa B/metabolismo , Invasividad Neoplásica , Pemetrexed , Neoplasias Pleurales/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Immunol ; 186(1): 32-40, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21106853

RESUMEN

IL-6 is a proinflammatory cytokine and its overproduction is implicated in a variety of inflammatory disorders. Recent in vitro analyses suggest that IL-6 is a key cytokine that determines the balance between Foxp3(+) regulatory T cells (Tregs) and Th17 cells. However, it remains unclear whether excessive IL-6 production in vivo alters the development and function of Foxp3(+) Tregs. In this study, we analyzed IL-6 transgenic (Tg) mice in which serum IL-6 levels are constitutively elevated. Interestingly, in IL-6 Tg mice, whereas peripheral lymphoid organs were enlarged, and T cells exhibited activated phenotype, Tregs were not reduced but rather increased compared with wild-type mice. In addition, Tregs from Tg mice normally suppressed proliferation of naive T cells in vitro. Furthermore, Tregs cotransferred with naive CD4 T cells into SCID-IL-6 Tg mice inhibited colitis as successfully as those transferred into control SCID mice. These results indicate that overproduction of IL-6 does not inhibit development or function of Foxp3(+) Tregs in vivo. However, when naive CD4 T cells alone were transferred, Foxp3(+) Tregs retrieved from SCID-IL-6 Tg mice were reduced compared with SCID mice. Moreover, the Helios(-) subpopulation of Foxp3(+) Tregs, recently defined as extrathymic Tregs, was significantly reduced in IL-6 Tg mice compared with wild-type mice. Collectively, these results suggest that IL-6 overproduced in vivo inhibits inducible Treg generation from naive T cells, but does not affect the development and function of natural Tregs.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/biosíntesis , Interleucina-6/biosíntesis , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/fisiología , Humanos , Interleucina-6/sangre , Interleucina-6/fisiología , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratones Transgénicos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/trasplante
9.
Int J Cancer ; 131(6): 1287-96, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22095154

RESUMEN

Inflammation is a crucial driving force in the development of gastric cancers (GCs). Accordingly, persistent activation of STAT3, a transcription factor pivotal in regulating both inflammation and oncogenesis, is often detected in GC, although its mechanism remains elusive. Suppressor of cytokine signaling-1 (SOCS-1) is a negative regulator of proinflammatory cytokine signaling and SOCS-1 gene methylation is frequently detected in various cancers including GC. However, the significance of SOCS-1 methylation in GC cells remains unexplored. Our study is undertaken to evaluate the role of SOCS-1 in GC cell proliferation and its effect on signaling pathways in GC cells. Among five GC cell lines, SOCS-1 gene was methylated in all cell lines and constitutive STAT3 phosphorylation with elevated endogenous IL-6 production was detected in two cell lines (NUGC-3 and AGS). Unexpectedly, anti-IL-6R antibody inhibited neither cell proliferation nor STAT3 phosphorylation in NUGC-3 and AGS. In contrast, enforced SOCS-1 expression by adenoviral vector (AdSOCS-1) markedly suppressed STAT3 phosphorylation and proliferation of NUGC-3 and AGS cells in vitro. Interestingly, the antiproliferative effect of SOCS-1 was attributable not only to the inhibition of STAT3 but also to that of p38 MAPK activity, and chemical inhibitors of JAK/STAT and p38 MAPK signaling effectively suppressed proliferation of these GC cells. Furthermore, treatment with AdSOCS-1 in vivo significantly suppressed GC proliferation in a xenograft model. These results suggest that SOCS-1 gene methylation is a critical step in the development of GC, and enforced expression of SOCS-1 may represent a novel therapeutic approach for the treatment of GC.


Asunto(s)
Factor de Transcripción STAT3/antagonistas & inhibidores , Neoplasias Gástricas/terapia , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular , Islas de CpG , Metilación de ADN , Activación Enzimática , Terapia Genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Fosfatidilinositol 3-Quinasas/fisiología , Receptores de Interleucina-6/fisiología , Factor de Transcripción STAT3/fisiología , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
10.
Int J Cancer ; 129(4): 1005-17, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20949562

RESUMEN

Malignant pleural mesothelioma (MPM) is an aggressive tumor with poor prognosis for which an effective therapy remains to be established. Our study investigated the therapeutic potential of the suppressor of cytokine signaling 3 (SOCS3), an endogenous inhibitor of intracellular signaling pathways, for treatment of MPM. We infected MPM cells (H226, EHMES-1, MESO-1 and MESO-4) with an adenovirus-expressing SOCS3 (AdSOCS3) to examine the effect of SOCS3 overexpression on MPM cells. SOCS3 overexpression reduced MPM proliferation and induced apoptosis and partial G0/G1 arrest. SOCS3 also inhibited the proliferation of MPM cells via multiple signaling pathways including Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase (ERK), focal adhesion kinase (FAK) and p53 pathways. Notably, AdSOCS3 treatment inhibited tumor growth in an MPM pleural xenograft model. These findings demonstrate that overexpression of SOCS3 has a potent antitumor effect against MPM both in vitro and in vivo and indicate the potential for clinical use of SOCS3 for MPM treatment.


Asunto(s)
Mesotelioma/prevención & control , Neoplasias Pleurales/prevención & control , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Adenoviridae/genética , Animales , Apoptosis , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Técnicas para Inmunoenzimas , Mesotelioma/metabolismo , Mesotelioma/patología , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int Immunol ; 22(5): 359-66, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20190037

RESUMEN

Therapeutic effects of green tea involve an inhibitory function of its constituent polyphenol epigallocatechin gallate (EGCG) on cell signaling. The specificity and mechanism(s) by which EGCG inhibits cell signaling have remained unclear. Here, we demonstrate that green tea and EGCG induce suppressor of cytokine signaling 1 (SOCS1) gene expression, a negative regulator of specific cell signaling pathways. In mouse immune cells, EGCG induces SOCS1 expression via an oxidative (superoxide) pathway and activation of the signal transducer and activator of transcription 5 transcription factor. EGCG inhibited SOCS1-regulated cell signaling, but this inhibitory effect was abrogated in cells deficient in SOCS1. These findings identify a mechanism by which EGCG inhibits cell signaling with specificity, mediated by induction of the negative regulator SOCS1.


Asunto(s)
Antioxidantes/farmacología , Catequina/análogos & derivados , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/genética , Té/química , Regulación hacia Arriba/efectos de los fármacos , Animales , Catequina/farmacología , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Relación Estructura-Actividad , Superóxidos/inmunología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/inmunología
12.
Proc Natl Acad Sci U S A ; 105(26): 9041-6, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18577591

RESUMEN

The development of Th17 cells is a key event in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine model of human multiple sclerosis (MS). Previous studies have demonstrated that an IL-6-dependent pathway is involved in the differentiation of Th17 cells from naïve CD4-positive T cells in vitro. However, the role of IL-6 in vivo in the development of Th17 cells in EAE has remained unclear. In the present study, we found that IL-6 blockade by treatment with an anti-IL-6 receptor monoclonal antibody (anti-IL-6R mAb) inhibited the development of EAE and inhibited the induction of myelin oligodendrocyte glycoprotein (MOG) peptide-specific CD4-positive, CD8-positive, and Th17 T cells, in inguinal lymph nodes. Thus, the protective effect of IL-6 blockade in EAE is likely to be mediated via the inhibition of the development of MOG-peptide-specific Th17 cells and Th1 cells, which in turn leads to reduced infiltration of T cells into the CNS. These findings indicate that anti-IL-6R mAb treatment might represent a novel therapy for human MS.


Asunto(s)
Antígenos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Epítopos/inmunología , Interleucina-6/inmunología , Vaina de Mielina/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Movimiento Celular , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Activación de Linfocitos/inmunología , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Péptidos/inmunología
13.
Int J Cancer ; 125(10): 2316-22, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19598262

RESUMEN

Clear cell carcinoma (CCC) of the ovary is known to be highly resistant to platinum-based chemotherapy. The purpose of our study was to identify a candidate protein that is associated with chemoresistance of CCC and to investigate the specific mechanism of chemoresistance conferred by the identified protein. Enhanced expression of Annexin A4 (Anx A4) was identified in ovarian CCC cells using 2-D differential gel electrophoresis (2D-DIGE) and mass spectrometry. Anx A4 levels were elevated in CCC cells compared with non-CCC cells as determined by real-time RT-PCR and Western blot analysis. Immunohistochemical analysis of Anx A4 was performed in 126 epithelial ovarian cancer tissue samples and demonstrated significantly elevated levels of Anx A4 protein levels in ovarian CCC tumors compared with ovarian serous and endometrioid tumors (p < 0.01). Anx A4-transfected ovarian non-CCC cells were more resistant to carboplatin (IC50 = 42 microM) compared with control cells (IC50 = 23 microM) as determined by modified MTT assay. Intracellular platinum levels were significantly lower in Anx A4-transfected cells compared with control cells after carboplatin treatment (p = 0.0020) and after an additional 360 min of carboplatin-free incubation (p = 0.0004), as measured by atomic absorption spectrophotometry. Expression of Anx A4 is elevated in ovarian CCC tumors and is associated with chemoresistance in cultured ovarian cancer cells. These results demonstrate that Anx A4 confers chemoresistance in ovarian cancer cells in part by enhancing drug efflux. Thus, Anx A4 may represent a novel therapeutic target of chemoresistance in patients with ovarian CCC.


Asunto(s)
Adenocarcinoma de Células Claras/metabolismo , Anexina A4/metabolismo , Antineoplásicos/uso terapéutico , Carboplatino/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias Ováricas/metabolismo , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/patología , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Electroforesis en Gel Bidimensional , Femenino , Humanos , Técnicas para Inmunoenzimas , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tasa de Supervivencia , Células Tumorales Cultivadas
14.
PLoS One ; 11(4): e0153399, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27070121

RESUMEN

BACKGROUND & AIM: Amyotrophic Lateral Sclerosis (ALS) is an adult-onset, progressive, motor neuron degenerative disease. Recent evidence indicates that inflammation is associated with many neurodegenerative diseases including ALS. Previously, abnormal levels of inflammatory cytokines including IL-1ß, IL-6 and TNF-α were described in ALS patients and/or in mouse ALS models. In addition, one study showed that blocking IL-1ß could slow down progression of ALS-like symptoms in mice. In this study, we examined a role for IL-6 in ALS, using an animal model for familial ALS. METHODS: Mice with mutant SOD1 (G93A) transgene, a model for familial ALS, were used in this study. The expression of the major inflammatory cytokines, IL-6, IL-1ß and TNF-α, in spinal cords of these SOD1 transgenic (TG) mice were assessed by real time PCR. Mice were then crossed with IL-6(-/-) mice to generate SOD1TG/IL-6(-/-) mice. SOD1 TG/IL-6(-/-) mice (n = 17) were compared with SOD1 TG/IL-6(+/-) mice (n = 18), SOD1 TG/IL-6(+/+) mice (n = 11), WT mice (n = 15), IL-6(+/-) mice (n = 5) and IL-6(-/-) mice (n = 8), with respect to neurological disease severity score, body weight and the survival. We also histologically compared the motor neuron loss in lumber spinal cords and the atrophy of hamstring muscles between these mouse groups. RESULTS: Levels of IL-6, IL-1ß and TNF-α in spinal cords of SOD1 TG mice was increased compared to WT mice. However, SOD1 TG/IL-6(-/-) mice exhibited weight loss, deterioration in motor function and shortened lifespan (167.55 ± 11.52 days), similarly to SOD1 TG /IL-6(+/+) mice (164.31±12.16 days). Motor neuron numbers and IL-1ß and TNF-α levels in spinal cords were not significantly different in SOD1 TG /IL-6(-/-) mice and SOD1 TG /IL-6 (+/+) mice. CONCLUSION: These results provide compelling preclinical evidence indicating that IL-6 does not directly contribute to motor neuron disease caused by SOD1 mutations.


Asunto(s)
Interleucina-6/deficiencia , Enfermedad de la Neurona Motora/genética , Neuronas Motoras/metabolismo , Mutación/genética , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Modelos Animales de Enfermedad , Femenino , Interleucina-1beta/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Enfermedad de la Neurona Motora/metabolismo , Médula Espinal/metabolismo , Superóxido Dismutasa-1 , Factor de Necrosis Tumoral alfa/genética
15.
J Exp Med ; 213(4): 605-19, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27022145

RESUMEN

Balance in signal transducer and activator of transcription (STAT) activation is a key factor in regulating the fate of naive CD4(+)T cells. Here, we demonstrate that AT-rich interactive domain-containing protein 5a (Arid5a) in T cells directs naive CD4(+)T cells to differentiate into inflammatory CD4(+)T cells, especially Th17 cells, through selective stabilization of Stat3(but not Stat1 and Stat5) mRNA in an IL-6-dependent manner. Loss of Arid5a in T cells led to reduction of STAT3 level under Th17-polarizing conditions, whereas STAT1 and STAT5 in Arid5a-deficient T cells were highly activated compared with those of WT T cells under the same conditions. These cells displayed the feature of antiinflammatory (Il10-expressing) CD4(+)T cells. Thus, we show a T cell-intrinsic role of Arid5a on fate decisions of naive CD4(+)T cells through selective stabilization of Stat3 mRNA.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Estabilidad del ARN/inmunología , ARN Mensajero/inmunología , Factor de Transcripción STAT3/inmunología , Células Th17/inmunología , Factores de Transcripción/inmunología , Animales , Proteínas de Unión al ADN/genética , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Noqueados , Estabilidad del ARN/genética , ARN Mensajero/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Células Th17/citología , Factores de Transcripción/genética
16.
Arthritis Rheum ; 58(12): 3710-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19035481

RESUMEN

OBJECTIVE: To investigate the mechanism of interleukin-6 (IL-6) blockade in autoimmune arthritis, by comparing the effect of anti-IL-6 receptor (anti-IL-6R) monoclonal antibody (mAb) treatment with the effect of soluble tumor necrosis factor (sTNFR)-Fc fusion protein treatment on T helper cell differentiation in collagen-induced arthritis (CIA). METHODS: DBA/1 mice were immunized with type II collagen (CII) to induce arthritis and were left untreated or were treated with anti-IL-6R mAb or TNFR-Fc. T helper cell differentiation and cytokine expression during the development of arthritis in these mice were analyzed. RESULTS: Immunization with CII predominantly increased the frequency of Th17 cells rather than Th1 cells. The frequency of FoxP3+ Treg cells was also increased after immunization. Treatment of mice with CIA with anti-IL-6R mAb on day 0 markedly suppressed the induction of Th17 cells and arthritis development, but treatment with this antibody on day 14 failed to suppress both Th17 differentiation and arthritis. In contrast, treatment of mice with CIA with TNFR-Fc from day 0 to day 14 suppressed neither Th17 differentiation nor arthritis, but treatment from day 21 to day 35 successfully ameliorated arthritis without inhibiting Th17 induction. Neither antibody treatment increased the frequency of Treg cells. CONCLUSION: Our results indicate that the protective effect of IL-6 blockade, but not tumor necrosis factor (TNF) blockade, in CIA correlates with the inhibition of Th17 differentiation. Our findings suggest that IL-6 blockade in rheumatoid arthritis in human is also likely to involve a therapeutic mechanism distinct from that of TNF blockade and thus may represent an alternative therapy for patients in whom the disease is refractory to TNF blockade.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Interleucina-6/antagonistas & inhibidores , Animales , Antirreumáticos/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Etanercept , Inmunoglobulina G/farmacología , Interleucina-17/inmunología , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Receptores de Interleucina-6/antagonistas & inhibidores , Receptores de Interleucina-6/inmunología , Receptores del Factor de Necrosis Tumoral , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA