Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 14(12): 1247-55, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24185614

RESUMEN

The inflammasome adaptor ASC contributes to innate immunity through the activation of caspase-1. Here we found that signaling pathways dependent on the kinases Syk and Jnk were required for the activation of caspase-1 via the ASC-dependent inflammasomes NLRP3 and AIM2. Inhibition of Syk or Jnk abolished the formation of ASC specks without affecting the interaction of ASC with NLRP3. ASC was phosphorylated during inflammasome activation in a Syk- and Jnk-dependent manner, which suggested that Syk and Jnk are upstream of ASC phosphorylation. Moreover, phosphorylation of Tyr144 in mouse ASC was critical for speck formation and caspase-1 activation. Our results suggest that phosphorylation of ASC controls inflammasome activity through the formation of ASC specks.


Asunto(s)
Proteínas del Citoesqueleto/inmunología , Inflamasomas/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas Quinasas JNK Activadas por Mitógenos/inmunología , Proteínas Tirosina Quinasas/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Células HEK293 , Humanos , Immunoblotting , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Nigericina/farmacología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Fosforilación/inmunología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Interferencia de ARN , Quinasa Syk , Tirosina/genética , Tirosina/inmunología , Tirosina/metabolismo
2.
PLoS Pathog ; 12(3): e1005507, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26991425

RESUMEN

Emergency myelopoiesis is inflammation-induced hematopoiesis to replenish myeloid cells in the periphery, which is critical to control the infection with pathogens. Previously, pro-inflammatory cytokines such as interferon (IFN)-α and IFN-γ were demonstrated to play a critical role in the expansion of hematopoietic stem cells (HSCs) and myeloid progenitors, leading to production of mature myeloid cells, although their inhibitory effects on hematopoiesis were also reported. Therefore, the molecular mechanism of emergency myelopoiesis during infection remains incompletely understood. Here, we clarify that one of the interleukin (IL)-6/IL-12 family cytokines, IL-27, plays an important role in the emergency myelopoiesis. Among various types of hematopoietic cells in bone marrow, IL-27 predominantly and continuously promoted the expansion of only Lineage-Sca-1+c-Kit+ (LSK) cells, especially long-term repopulating HSCs and myeloid-restricted progenitor cells with long-term repopulating activity, and the differentiation into myeloid progenitors in synergy with stem cell factor. These progenitors expressed myeloid transcription factors such as Spi1, Gfi1, and Cebpa/b through activation of signal transducer and activator of transcription 1 and 3, and had enhanced potential to differentiate into migratory dendritic cells (DCs), neutrophils, and mast cells, and less so into macrophages, and basophils, but not into plasmacytoid DCs, conventional DCs, T cells, and B cells. Among various cytokines, IL-27 in synergy with the stem cell factor had the strongest ability to augment the expansion of LSK cells and their differentiation into myeloid progenitors retaining the LSK phenotype over a long period of time. The experiments using mice deficient for one of IL-27 receptor subunits, WSX-1, and IFN-γ revealed that the blood stage of malaria infection enhanced IL-27 expression through IFN-γ production, and the IL-27 then promoted the expansion of LSK cells, differentiating and mobilizing them into spleen, resulting in enhanced production of neutrophils to control the infection. Thus, IL-27 is one of the limited unique cytokines directly acting on HSCs to promote differentiation into myeloid progenitors during emergency myelopoiesis.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Interleucinas/metabolismo , Mielopoyesis/fisiología , Animales , Linfocitos B/efectos de los fármacos , Médula Ósea/fisiología , Diferenciación Celular , Linaje de la Célula , Citocinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/fisiología , Células Progenitoras Mieloides/fisiología , Transducción de Señal , Bazo/fisiología
3.
Mol Syst Biol ; 12(3): 861, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27013061

RESUMEN

Epigenetic mechanisms have emerged as links between prenatal environmental exposure and disease risk later in life. Here, we studied epigenetic changes associated with maternal smoking at base pair resolution by mapping DNA methylation, histone modifications, and transcription in expectant mothers and their newborn children. We found extensive global differential methylation and carefully evaluated these changes to separate environment associated from genotype-related DNA methylation changes. Differential methylation is enriched in enhancer elements and targets in particular "commuting" enhancers having multiple, regulatory interactions with distal genes. Longitudinal whole-genome bisulfite sequencing revealed that DNA methylation changes associated with maternal smoking persist over years of life. Particularly in children prenatal environmental exposure leads to chromatin transitions into a hyperactive state. Combined DNA methylation, histone modification, and gene expression analyses indicate that differential methylation in enhancer regions is more often functionally translated than methylation changes in promoters or non-regulatory elements. Finally, we show that epigenetic deregulation of a commuting enhancer targeting c-Jun N-terminal kinase 2 (JNK2) is linked to impaired lung function in early childhood.


Asunto(s)
Epigénesis Genética , Secuencias Reguladoras de Ácidos Nucleicos , Fumar/genética , Niño , Cromatina/metabolismo , Estudios de Cohortes , Metilación de ADN , Femenino , Histonas/metabolismo , Humanos , Masculino , Proteína Quinasa 9 Activada por Mitógenos/genética , Madres , Fenotipo , Polimorfismo de Nucleótido Simple , Transcripción Genética
4.
Cancer Sci ; 104(9): 1146-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23758044

RESUMEN

A number of CML patients who achieve a sustained complete molecular response (CMR) for at least 2 years during imatinib (IM) therapy can discontinue IM without relapse. With the long-term goal of developing immunological criteria for managing IM therapy in CML patients, we compared the immunophenotypic profiles of three groups of CML patients: those who received IM and had a CMR for more than two consecutive years (CMR group); patients who received IM and did not have a sustained CMR but maintained a major molecular response for more than 2 years (fluctuating CMR group); and patients with a sustained CMR for more than 6 months after IM discontinuation (STOP-IM group), together with healthy controls. The percentages of effector populations of natural killer (NK) cells, such as interferon (IFN)-γ(+) CD3(-) CD56(+) cells, were significantly higher in the STOP-IM and CMR groups than in the fluctuating CMR and control groups. The elevated levels of these effector NK cells were sustained for more than 3 years after IM discontinuation. In contrast, the percentages of effector memory CD8(+) T cells, such as IFN-γ(+) CCR7(-) CD45RO(+) CD8(+) cells, were significantly higher in the STOP-IM and control groups than in the CMR and fluctuating CMR groups, possibly owing to IM intake. These results suggest that the immunological activation status of NK cells contributes to CMR maintenance. Higher activation levels of effector NK cells in CML patients being treated with IM might reflect minimization of BCR-ABL1 transcript levels and therefore could be additive information for determining whether to stop IM.


Asunto(s)
Benzamidas/uso terapéutico , Células Asesinas Naturales/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Linfocitos T CD8-positivos/metabolismo , Femenino , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba
5.
Int Arch Allergy Immunol ; 162(2): 104-14, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23921222

RESUMEN

BACKGROUND: Bronchial asthma is a chronic inflammatory disease of the airway. Apoptosis signal-regulating kinase 1 (ASK1), a member of the mitogen-activated protein kinase kinase kinase family, is activated by environmental stress and plays a crucial role in the induction of apoptosis and inflammation. To examine whether ASK1 is involved in the induction of bronchial asthma, we investigated the role of ASK1 using a genetic approach in the production of cytokines, as well as the development of airway hyperreactivity (AHR) and antibody responses using a murine airway inflammation model. METHODS: ASK1-deficient (ASK1(-/-)) and control wild-type (WT) mice were immunized with ovalbumin (OVA) without alum intraperitoneally, followed by intranasal administration of OVA. Airway infiltration of inflammatory cells, cytokine production, AHR and antibody production were assayed. The asthmatic phenotype was assessed following intranasal administration of IL-13 or TNF-α. RESULTS: ASK1(-/-) mice sensitized with OVA displayed an impaired inflammatory cell infiltration into airways and a decreased AHR relative to WT mice. Moreover, the production of OVA-specific IgE antibodies and proasthmatic cytokines (IL-5, IL-13 and TNF-α) was substantially reduced in OVA-stimulated ASK1(-/-) mice. Intranasal administration of IL-13 and OVA enhanced the accumulation of inflammatory cells in OVA-primed ASK1(-/-) mice. The OVA-induced AHR in response to methacholine was enhanced by IL-13 in WT mice but not ASK1(-/-) mice. CONCLUSIONS: The ASK1 signaling pathway regulates the OVA-induced asthmatic phenotype, specifically AHR sensitivity and cytokine production. Therefore, the ASK1 signaling pathway is a promising target for therapeutic intervention in some asthmatic patients.


Asunto(s)
Asma/inmunología , Hiperreactividad Bronquial/inmunología , Pulmón/inmunología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Animales , Apoptosis , Asma/metabolismo , Hiperreactividad Bronquial/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , Eosinófilos/inmunología , Células Caliciformes/inmunología , Inmunoglobulina E/inmunología , Inflamación , Interleucina-13/administración & dosificación , Interleucina-13/biosíntesis , Interleucina-13/metabolismo , Interleucina-5/biosíntesis , Pulmón/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Sistema de Señalización de MAP Quinasas , Cloruro de Metacolina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Ovalbúmina , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
6.
Microbiol Immunol ; 57(3): 185-92, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23278594

RESUMEN

Protein arginine methylation plays crucial roles, including signal transduction, transcriptional control, cell proliferation and/or differentiation. B cells undergo clonal division, isotype switching and differentiate into antibody forming cells following stimulation with Toll-like receptor-ligand, lipopolysaccharide (LPS) and T cell-derived signals, including CD40-ligand (CD40-L) and interleukin 4 (IL-4). Whether protein arginine methylation affects B cell division and/or isotype switching to IgG1 in response to LPS, IL-4, and CD40-L was examined using the arginine methyl transferase inhibitor adenosine-2',3'-dialdehyde (AdOx). Addition of AdOx substantially reduced the number of division cycles of stimulated B cells, whereas cell viability remained intact. Upon stimulation with LPS/IL-4/CD40-L, the proportion of surface IgG1 positive cells in each division cycle was slightly diminished by AdOx. However, the degree of expression of γ1 germ line transcript and activation-induced cytidine deaminase (AID) in response to LPS/IL-4/CD40-L were unaffected by addition of AdOx, suggesting that AdOx influences class switch recombination independent of AID expression through transcriptional control. Taken together, arginine methylation appears to be involved in B cell isotype switching, as well as in clonal expansion of B cells in response to LPS/IL-4/CD40-L.


Asunto(s)
Arginina/metabolismo , Linfocitos B/inmunología , División Celular , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/biosíntesis , Procesamiento Proteico-Postraduccional , Animales , Ligando de CD40/inmunología , Proliferación Celular , Interleucina-4/inmunología , Lipopolisacáridos/inmunología , Metilación , Ratones , Ratones Endogámicos C57BL
7.
Clin Dev Immunol ; 2013: 968549, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23956763

RESUMEN

T-helper 17 (Th17) cells are characterized by producing interleukin-17 (IL-17, also called IL-17A), IL-17F, IL-21, and IL-22 and potentially TNF- α and IL-6 upon certain stimulation. IL-23, which promotes Th17 cell development, as well as IL-17 and IL-22 produced by the Th17 cells plays essential roles in various inflammatory diseases, such as experimental autoimmune encephalomyelitis, rheumatoid arthritis, colitis, and Concanavalin A-induced hepatitis. In this review, we summarize the characteristics of the functional role of Th17 cells, with particular focus on the Th17 cell-related cytokines such as IL-17, IL-22, and IL-23, in mouse models and human inflammatory diseases.


Asunto(s)
Citocinas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Animales , Humanos , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Interleucinas/metabolismo , Interleucina-22
8.
BMC Cell Biol ; 13: 7, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22435755

RESUMEN

BACKGROUND: Interferon-α (IFN-α) exerts an anti-tumor effect at least through induction of apoptosis in a variety of types including B lymphoma cells. We recently found that IFN-α induced a sustained activation of c-Jun NH2-terminal kinase1 (JNK1), which is implicated in activation of the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) promoter. In the present study, we explored upstream component(s) of the prolonged IFN-α-initiated activation of JNK1. RESULTS: IFN-α caused activation of PKC-δ in Daudi B lymphoma cells and myeloma U266 cells, as detected by Western blotting using a monoclonal antibody specific for the phosphorylated form of PKC-δ. The dominant-negative form of mutant PKC-δ (dnPKC-δ) reduced the IFN-α-induced JNK1 activation, TRAIL promoter activity, loss of mitochondrial membrane potential (ΔΨm), and increase in propidium iodide (PI) positive cells. The IFN-α-induced activation of JNK1 and the TRAIL promoter was also attenuated by the PKC-δ inhibitor rottlerin. Moreover, a constitutively active form of mutant PKC-δ enhanced the IFN-α-induced TRAIL promoter activity and loss of ΔΨm in Daudi B lymphoma cells. In addition, IFN-α-induced Ser727 phosphorylation of Stat1 was also abrogated by dnPKC-δ. CONCLUSIONS: IFN-α induced JNK1 activation via PKC-δ, leading to upregulation of TRAIL. The interaction of the consequent enhanced TRAIL expression with TRAIL-receptor results in a loss of ΔΨm and increase in PI positive cells. The IFN-α-induced apoptotic events may also be affected by the Ser727-Stat1 induced by PKC-δ-mediated signaling component(s).


Asunto(s)
Apoptosis , Interferón-alfa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma de Células B/enzimología , Proteína Quinasa C-delta/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Linfoma de Células B/fisiopatología , Regiones Promotoras Genéticas , Proteína Quinasa C-delta/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
9.
Eur J Immunol ; 41(10): 2828-39, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21953641

RESUMEN

IL-23 plays a critical role in the expansion of highly proinflammatory Th17 cells secreting IL-17 and IL-22. Recently, we demonstrated that Notch signaling drives IL-22 secretion through the aryl hydrocarbon receptor (AHR) and plays a protective role in Con A-induced hepatitis. In this study, we investigated the role of IL-23 in hepatitis using IL-23p19- and IL-17-deficient mice. In WT mice, the injection of Con A induced the upregulation of various cytokines, which included IL-23, IL-22, IL-17, IFN-γ and TNF-α. In IL-23p19-deficient mice, exacerbated hepatitis was observed and serum IL-22 and IL-17 levels were greatly reduced, whereas in IL-17-deficient mice, ameliorated hepatitis was observed. The injection of exogenous IL-22 protected p19-deficient mice from hepatitis, whereas the injection of exogenous IL-23 significantly increased the serum levels of not only IL-22 but also IL-17, and less effectively protected against hepatitis in IL-17-dependent and -independent manners. Finally, it was revealed that STAT3, STAT4 and Notch contributed to the production of both the cytokines, and that the AHR was important only for IL-22 production in response to Con A and IL-23 in liver mononuclear cells. These results suggest that IL-23 plays a protective role in hepatitis through IL-22 production and also a pathological role via IL-17-dependent and -independent mechanisms.


Asunto(s)
Hepatitis Animal/inmunología , Hepatitis Animal/metabolismo , Interleucina-17/metabolismo , Subunidad p19 de la Interleucina-23/metabolismo , Interleucina-23 , Interleucinas/metabolismo , Animales , Concanavalina A , Citocinas/biosíntesis , Interleucina-17/sangre , Interleucina-17/genética , Interleucina-23/administración & dosificación , Interleucina-23/biosíntesis , Interleucina-23/metabolismo , Interleucina-23/farmacología , Subunidad p19 de la Interleucina-23/genética , Interleucinas/administración & dosificación , Interleucinas/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Hidrocarburo de Aril/metabolismo , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT4/metabolismo , Transducción de Señal/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Interleucina-22
10.
Cell Mol Biol Lett ; 17(1): 36-48, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22139584

RESUMEN

Thy28 protein is conserved among plants, bacteria, and mammalian cells. Nuclear Thy28 protein is substantially expressed in testis, liver, and immune cells such as lymphocytes. Lymphocyte apoptosis plays a crucial role in homeostasis and formation of a diverse lymphocyte repertoire. In this study, we examined whether Thy28 affects induction of apoptosis in WEHI-231 B lymphoma cells following engagement of membrane immunoglobulin (mIg). Once they were established, the Thy28-overexpressing WEHI-231 cells showed similar expression levels of IgM and class I major histocompatibility complex (MHC) molecule compared with controls. The Thy28-overexpressing cells were considerably resistant to loss of mitochondrial membrane potential (ΔΨm), caspase-3 activation, and increase in annexin-positive cells upon mIg engagement. These changes were concomitant with an increase in G1 phase associated with upregulation of p27(Kip1). The anti-IgM-induced sustained activation of c-Jun N-terminal kinase (JNK), which was associated with late-phase hydrogen peroxide (H(2)O(2)) production, was partially reduced in the Thy28-expressing cells relative to controls. Taken together, the data suggest that in WEHI-231 B lymphoma cells, Thy28 regulates mIg-mediated apoptotic events through the JNK-H(2)O(2) activation pathway, concomitant with an accumulation of cells in G1 phase associated with upregulation of p27(Kip1) in WEHI-231 B lymphoma cells.


Asunto(s)
Apoptosis , Inmunoglobulinas/metabolismo , Proteínas Nucleares/metabolismo , Animales , Caspasa 3/metabolismo , Membrana Celular/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Fase G1 , Genes MHC Clase I/genética , Peróxido de Hidrógeno/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Células Tumorales Cultivadas , Regulación hacia Arriba
11.
J Biomed Biotechnol ; 2010: 605483, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20454646

RESUMEN

Cytotoxic T lymphocytes (CTLs) play a critical role in the control of various cancers and infections, and therefore the molecular mechanisms of CTL generation are a critical issue in designing antitumor immunotherapy and vaccines which augment the development of functional and long-lasting memory CTLs. Interleukin (IL)-27, a member of the IL-6/IL-12 heterodimeric cytokine family, acts on naive CD4+ T cells and plays pivotal roles as a proinflammatory cytokine to promote the early initiation of type-1 helper differentiation and also as an antiinflammatory cytokine to limit the T cell hyperactivity and production of pro-inflammatory cytokines. Recent studies revealed that IL-27 plays an important role in CD8+ T cells as well. Therefore, this article reviews current understanding of the role of IL-27 in CD8+ T cell functions and generation of CTLs.


Asunto(s)
Interleucina-17/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Animales , Humanos , Interferón gamma/biosíntesis , Neoplasias/inmunología , Especificidad de Órganos/inmunología , Transducción de Señal/inmunología
12.
Clin Dev Immunol ; 20102010.
Artículo en Inglés | MEDLINE | ID: mdl-20885915

RESUMEN

The interleukin (IL)-12 family, which is composed of heterodimeric cytokines including IL-12, IL-23, and IL-27, is produced by antigen-presenting cells such as macrophages and dendritic cells and plays critical roles in the regulation of helper T (Th) cell differentiation. IL-12 induces IFN-γ production by NK and T cells and differentiation to Th1 cells. IL-23 induces IL-17 production by memory T cells and expands and maintains inflammatory Th17 cells. IL-27 induces the early Th1 differentiation and generation of IL-10-producing regulatory T cells. In addition, these cytokines induce distinct immune responses to tumors. IL-12 activates signal transducers and activator of transcription (STAT)4 and enhances antitumor cellular immunity through interferon (IFN)-γ production. IL-27 activates STAT1, as does IFN-γ and STAT3 as well, and enhances antitumor immunity by augmenting cellular and humoral immunities. In contrast, although exogenously overexpressed IL-23 enhances antitumor immunity via memory T cells, endogenous IL-23 promotes protumor immunity through STAT3 activation by inducing inflammatory responses including IL-17 production.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Interleucina-12/inmunología , Neoplasias/inmunología , Diferenciación Celular , Regulación de la Expresión Génica/inmunología , Humanos , Interleucina-12/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-23/inmunología , Interleucina-23/metabolismo , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología
13.
Exp Cell Res ; 315(20): 3467-76, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19766627

RESUMEN

Engagement of membrane immunoglobulin (mIg) on WEHI-231 mouse B lymphoma cells results in growth arrest at the G1 phase of the cell cycle, followed by a reduction of mitochondrial membrane potential (DeltaPsim) and apoptosis. WEHI-231 cells resemble immature B cells in terms of the cell surface phenotype and sensitivity to mIg engagement. However, the molecular mechanisms underlying mIg-induced loss of DeltaPsim and apoptosis have not yet been established. In this study, we show that apoptosis signal-regulating kinase 1 (ASK1)-c-Jun N-terminal kinase 1 (JNK1) signaling pathway participates in mIg-induced apoptosis through the generation of reactive oxygen species (ROS). Stimulation of WEHI-231 cells with anti-IgM induces phosphorylation and subsequent activation of ASK1, leading to JNK activation. Anti-IgM stimulation immediately (5 min) induces hydrogen peroxide (H2O2) production with a substantial increase during later time points (36-48 h), accompanied by loss of DeltaPsim and an increase in cells with sub-G1 DNA content. The anti-IgM-induced late-phase H2O2 production, loss of DeltaPsim, and increase in the sub-G1 fraction were all reduced substantially in WEHI-231 cells overexpressing a dominant-negative form of ASK1, compared with control vector alone, but enhanced substantially in cells overexpressing a constitutively active form of ASK1. These mIg-mediated events were also partially abrogated by ROS scavenger N-acetyl-L-cysteine (NAC). Taken together, these results suggest that mIg engagement induces H2O2 production leading to activation of ASK1-JNK1 pathway, creating a feedback amplification loop of ROS-ASK/JNK that leads to loss of DeltaPsim and finally apoptosis.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Acetilcisteína/farmacología , Animales , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Antiidiotipos/farmacología , Apoptosis/genética , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Línea Celular Tumoral , Citosol/metabolismo , Activación Enzimática , Fase G1/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Transporte de Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Proteína X Asociada a bcl-2/metabolismo
14.
Biochem Biophys Res Commun ; 386(1): 105-10, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19501566

RESUMEN

TGF-beta and IL-6 induce Th17 differentiation, and IL-23 is required for expansion and maintenance of Th17 cells. Recently, it was shown that IL-6 up-regulates IL-23R mRNA in naive CD4+ T cells and therefore IL-6 and IL-23 synergistically promote Th17 differentiation. However, the molecular mechanism whereby IL-6 and IL-23 induce Th17 differentiation and the relevance to TGF-beta remain unknown. Here, we found that IL-6 up-regulated IL-23R mRNA expression, and IL-6 and IL-23 synergistically augmented its protein expression. The combination induced Th17 differentiation, and TGF-beta1 further enhanced it. IL-6 augmented endogenous TGF-beta1 mRNA expression, whereas the amount of TGF-beta produced was not enough to induce Th17 differentiation by IL-6 alone. However, unexpectedly, the up-regulation of IL-23R and induction of Th17 differentiation by IL-6 and IL-23 were almost completely inhibited by anti-TGF-beta. These results suggest that the induction of IL-23R and Th17 differentiation by IL-6 and IL-23 is mediated through endogenously produced TGF-beta.


Asunto(s)
Diferenciación Celular , Interleucina-17/inmunología , Receptores de Interleucina/biosíntesis , Linfocitos T Colaboradores-Inductores/inmunología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Células Cultivadas , Interleucina-23/inmunología , Interleucina-23/farmacología , Interleucina-6/inmunología , Interleucina-6/farmacología , Ratones , Ratones Mutantes , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos
15.
Oncol Rep ; 22(5): 1169-72, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19787236

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces apoptosis in MIT6 cells derived from primary oral squamous cell carcinoma (OSCC), whereas it does not induce apoptosis in MIL6 cells derived from metastases. The present studies were performed to examine whether activation of c-Jun NH2-terminal kinase (JNK) is implicated in the differential sensitivity to TRAIL-induced apoptosis. The TRAIL-induced JNK activation in MIT6 cells was stronger than in MIL6 cells, as assessed by Western blotting using antibodies specific for phospho-JNK. To evaluate the role of JNK1 in TRAIL-induced cell death, one clone expressing the dominant-negative form of JNK1 (dnJNK1) was established. The dnJNK1-expressing cells and MIL6 cells expressed TRAIL protein at levels similar to or even greater than MIT6 cells did. When cell death was assessed by annexin V staining and mitochondrial membrane potential, kinetic studies demonstrated that the dnJNK1-expressing cells were substantially more resistant to 100 ng/ml TRAIL, comparable to MIL6 cells, at 36 and 48 h after stimulation. Collectively, the primary OSCC cell line, MIT6, is sensitive to TRAIL but its metastatic line MIL6 is resistant to TRAIL exposure. Thus, the underlying molecular mechanism of TRAIL-induced cell death involves JNK activation. These results suggest that the acquisition of TRAIL resistance provides some metastatic capacity to primary tumors.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias de la Boca/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Western Blotting , Resistencia a Antineoplásicos , Activación Enzimática , Citometría de Flujo , Genes Dominantes , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Potencial de la Membrana Mitocondrial , Células Tumorales Cultivadas
16.
J Cell Biochem ; 104(5): 1927-36, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18384124

RESUMEN

WEHI-231 B lymphoma cells have been employed for analysis of antigen-induced B cell unresponsiveness because these cells undergo cell cycle arrest in G1, accompanied by induction of apoptosis. In the present study, we examined the requirement for toxic small molecules apoptosis-inducing factor (AIF) and cytochrome c, and subsequent caspase activation in apoptotic cell death in WEHI-231 and CH31 B lymphoma cells following engagement of membrane immunoglobulin (mIg). Pan-caspase inhibitor BD-fmk blocked mIg-mediated increase in cells with sub-G1 DNA content, whereas it did not affect mIg-mediated loss of mitochondrial membrane potential and phosphatidylserine exposure on B cell membrane. Dominant-negative form of c-Jun NH2-terminal kinase1 (JNK1) blocked the translocation of AIF into the nuclei and cytosol from the mitochondria in the WEHI-231 and CH31 cells following mIg engagement, whereas constitutively active form of JNK1 enhanced it. This AIF translocation was also blocked by Bcl-xL, but not by BD-fmk. Moreover, AIF-deficient clones via small interfering RNA (siRNA)-mediated method showed small increase in loss of mitochondrial membrane potential. After mIg engagement, the AIF-deficient clones displayed an enhanced sensitivity to mIg-mediated apoptosis, concomitant with translocation of a residual AIF into the nuclei, compared with control clone. Our findings are compatible with the notion that AIF has dual role, with a proapoptotic function in the nuclei and a distinct anti-apoptotic function in the mitochondria. These observations would be valuable for analysis of B cell unresponsiveness and hopefully for treatment of diseases involving B cell dysfunction.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Inmunoglobulina M/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma/enzimología , Linfoma/patología , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Células Clonales , Citosol/efectos de los fármacos , Citosol/metabolismo , ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Fase G1/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosfatidilserinas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteína bcl-X/metabolismo
17.
Cancer Chemother Pharmacol ; 62(4): 569-76, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18026729

RESUMEN

PURPOSE: Exposure to carboplatin (CBDCA) has been demonstrated to result in apoptotic and/or necrotic cell death, but molecular mechanisms underlying CBDCA-induced apoptosis or necrosis remain largely unclear. Here, we examined whether activation of c-Jun NH(2)-terminal kinase (JNK) modulates the mode of cell death induced by CBDCA in CD31 B lymphoma cells. METHODS: The mode of cell death (apoptosis versus necrosis) was investigated by flow cytometry using 7-amino-actinomycin D (7-AAD) and annexin-FITC probes. To evaluate the role of JNK1 in CBDCA-induced cell death, CH31 B lymphoma cells overexpressing dominant-negative form of JNK1 (dnJNK1) or constitutively active form of JNK1 (MKK7-JNK1) were established. Intracellular accumulation of superoxide anion (O(2) (-)) was determined by flow cytometry using the fluorescent probe dihydroethidium (DHE). RESULTS: The CBDCA-induced primary apoptosis and secondary necrosis were abrogated in the dnJNK1-overexpressing CH31 cells, while it was somewhat enhanced in the MKK7-JNK1-overexpressing cells. In contrast, the CBDCA-induced primary necrosis was reduced by MKK7-JNK1, with a concurrent decrease in production of O(2) (-). The superoxide anion scavenger for butylated hydroxyanisol (BHA) partially reduced the CBDCA-induced O(2) (-) production and necrotic, but not apoptotic, death in both wild type and dnJNK1-overexpressing CH31 cells. CONCLUSIONS: Prolonged activation of JNK1 appears to be involved in CBDCA-induced apoptosis with prevention of necrosis induction, and the induction of necrosis appears to correlate with CBDCA-induced O(2) (-) production, which is partially blocked by co-culture with BHA. These observations provide valuable information for understanding molecular mechanisms underlying CBDCA-induced cell death, and hopefully for the design of novel treatment modalities for patients with tumors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carboplatino/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma de Células B , MAP Quinasa Quinasa 7/metabolismo , Animales , Hidroxianisol Butilado/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Activación Enzimática/efectos de los fármacos , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/enzimología , Linfoma de Células B/patología , Ratones , Necrosis/inducido químicamente , Superóxidos/metabolismo
18.
Oncol Rep ; 18(4): 1031-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17786370

RESUMEN

IFN-alpha regulates tumor cell growth at least through induction of apoptosis. We have recently demonstrated that IFN-alpha causes apoptosis through upregulation of TNF-related apoptosis-inducing ligand (TRAIL) in Daudi B lymphoma and U266 myeloma cells. However, other cell lines such as Ramos and RPMI 8226 underwent apoptosis without any apparent involvement of TRAIL following IFN-alpha stimulation. In this study, we examined whether the IFN-alpha-induced upregulation of TRAIL is essential for the induction of apoptosis. IFN-alpha-induced early phase (48 h) of loss of DeltaPsim was substantially prevented in Daudi B lymphoma cells overexpressing the dominant-negative form of Fas-associated death domain (dnFADD) compared with vector control, whereas a late phase (72 h) of DeltaPsim was comparable to the control. The IFN-alpha-induced early phase of apoptosis was also reduced in the dnFADD-expressing cells, while the late phase of apoptosis was unaffected. IFN-alpha-induced upregulation of TRAIL protein in the dnFADD-expressing Daudi or U266 cells was comparable to their control cells, suggesting that FADD is not involved in the IFN-alpha-induced upregulation of TRAIL. Moreover, the early phase of mitochondrial depolarization was severely prevented by the presence of fusion protein of TRAIL receptor 1 and Fc portion of immunoglobulin (TRAIL-R1:Fc) and TRAIL-R2:Fc. Together, IFN-alpha induces apoptosis in a TRAIL-dependent or -independent manner, depending on the course of the apoptotic process.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Interferón-alfa/farmacología , Linfoma de Células B/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/inmunología , Western Blotting , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Citometría de Flujo , Genes Dominantes , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Fracciones Subcelulares , Células Tumorales Cultivadas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
19.
J Interferon Cytokine Res ; 26(6): 421-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16734563

RESUMEN

Interferon-alpha (IFN-alpha) regulates multiple biologic functions, including antiviral activity, immune regulation, cell differentiation, and cell survival or death, in a variety of cell types. We and others have recently demonstrated that IFN-alpha induces cell death through activation of c-Jun NH(2)-terminal kinase (JNK) in human Daudi B lymphoma and U266 myeloma cells. Moreover, the IFN-alpha-induced signaling pathway has been shown to cross talk with the antigen receptor-mediated signaling cascade. In the present study, we examined whether IFN-alpha affects cell death after engagement of membrane immunoglobulin (mIg) using anti-IgM. Daudi cells pretreated with low concentrations of IFN-alpha (25 or 250 U/mL) for 24 h were stimulated with anti-IgM (1-10 microg/mL) for 24 h. The cells were assayed for JNK activation, mitochondrial membrane potential (DeltaPsim) by Western blotting, and DiOC(6) staining, respectively. The IFN-alpha-primed Daudi cells showed an increased sensitivity to subsequent stimulation with anti-IgM, as assessed by JNK activation and DeltaPsim. Moreover, Daudi cells overexpressing the constitutively active or dominant-negative form of JNK were substantially susceptible or resistant to anti-IgM-induced DeltaPsim, respectively, compared with cells overexpressing the control vector alone. Taken together, these results indicate that IFN-alpha renders Daudi B lymphoma cells susceptible to anti-IgM-induced apoptosis, probably through upregulation of JNK activation.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Interferón-alfa/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma de Células B/metabolismo , Mitocondrias/fisiología , Membranas Mitocondriales/fisiología , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Activación Enzimática/efectos de los fármacos , Genes Reporteros , Humanos , Luciferasas/metabolismo , Linfoma de Células B/tratamiento farmacológico , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos
20.
J Orthop Res ; 24(6): 1153-62, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705697

RESUMEN

Molecular mechanisms underlying chemotherapeutic agent-induced apoptosis in sarcoma cells are not well known. Induction of apoptosis is regulated by several components including mitogen-activated protein kinases (MAPKs) comprising ERK, p38MAPKs, and c-Jun N-terminal kinase (JNK). In the present study, we examined whether activation of JNK is induced by the chemotherapeutic agents cis-diaminedichloroplatinum (cisplatin, CDDP) or doxorubicin (DXR), and whether the ectopic expression of constitutively active (MKK7-JNK1) or dominant-negative form of JNK (dnJNK) influenced apoptosis in response to the CDDP or DXR in sarcoma cell lines MG-63 and SaOS-2. The CDDP or DXR induced JNK activation in the both cell lines, as assessed by Western blotting using phosphospecific antibodies. A transient expression of the activated form of JNK sensitized the MG-63 and SaOS-2 cells to the drug-induced apoptosis, while dnJNK1 reduced the proportion of apoptotic cell death. Apoptosis was determined by flow cytometry using annexin-V Cy5. Collectively, our results indicate that JNK activation is involved in apoptotic cell death in sarcoma cell lines following stimulation with CDDP or DXR.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Doxorrubicina/farmacología , Proteína Quinasa 8 Activada por Mitógenos/biosíntesis , Sarcoma/tratamiento farmacológico , Western Blotting , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Citometría de Flujo , Humanos , Isoenzimas , Sarcoma/enzimología , Sarcoma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA