Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 2(1): 100188, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33521702

RESUMEN

Chordomas are rare spinal tumors addicted to expression of the developmental transcription factor brachyury. In chordomas, brachyury is super-enhancer associated and preferentially downregulated by pharmacologic transcriptional CDK inhibition, leading to cell death. To understand the underlying basis of this sensitivity, we dissect the brachyury transcription regulatory network and compare the consequences of brachyury degradation with transcriptional CDK inhibition. Brachyury defines the chordoma super-enhancer landscape and autoregulates through binding its super-enhancer, and its locus forms a transcriptional condensate. Transcriptional CDK inhibition and brachyury degradation disrupt brachyury autoregulation, leading to loss of its transcriptional condensate and transcriptional program. Compared with transcriptional CDK inhibition, which globally downregulates transcription, leading to cell death, brachyury degradation is much more selective, inducing senescence and sensitizing cells to anti-apoptotic inhibition. These data suggest that brachyury downregulation is a core tenet of transcriptional CDK inhibition and motivates developing strategies to target brachyury and its autoregulatory feedback loop.


Asunto(s)
Biomarcadores de Tumor/genética , Cordoma/genética , Quinasas Ciclina-Dependientes/genética , Proteínas Fetales/genética , Proteínas de Neoplasias/genética , Neoplasias de la Columna Vertebral/genética , Proteínas de Dominio T Box/genética , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Cordoma/metabolismo , Cordoma/patología , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Fetales/metabolismo , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Queratina-18/genética , Queratina-18/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas de Neoplasias/metabolismo , Proteolisis , Transducción de Señal , Neoplasias de la Columna Vertebral/metabolismo , Neoplasias de la Columna Vertebral/patología , Proteínas de Dominio T Box/metabolismo
2.
Nat Med ; 25(2): 292-300, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30664779

RESUMEN

Chordoma is a primary bone cancer with no approved therapy1. The identification of therapeutic targets in this disease has been challenging due to the infrequent occurrence of clinically actionable somatic mutations in chordoma tumors2,3. Here we describe the discovery of therapeutically targetable chordoma dependencies via genome-scale CRISPR-Cas9 screening and focused small-molecule sensitivity profiling. These systematic approaches reveal that the developmental transcription factor T (brachyury; TBXT) is the top selectively essential gene in chordoma, and that transcriptional cyclin-dependent kinase (CDK) inhibitors targeting CDK7/12/13 and CDK9 potently suppress chordoma cell proliferation. In other cancer types, transcriptional CDK inhibitors have been observed to downregulate highly expressed, enhancer-associated oncogenic transcription factors4,5. In chordoma, we find that T is associated with a 1.5-Mb region containing 'super-enhancers' and is the most highly expressed super-enhancer-associated transcription factor. Notably, transcriptional CDK inhibition leads to preferential and concentration-dependent downregulation of cellular brachyury protein levels in all models tested. In vivo, CDK7/12/13-inhibitor treatment substantially reduces tumor growth. Together, these data demonstrate small-molecule targeting of brachyury transcription factor addiction in chordoma, identify a mechanism of T gene regulation that underlies this therapeutic strategy, and provide a blueprint for applying systematic genetic and chemical screening approaches to discover vulnerabilities in genomically quiet cancers.


Asunto(s)
Cordoma/metabolismo , Proteínas Fetales/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Proliferación Celular/efectos de los fármacos , Cordoma/genética , Cordoma/patología , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Genes Esenciales , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología
3.
Nat Commun ; 8(1): 890, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29026114

RESUMEN

Chordoma is a malignant, often incurable bone tumour showing notochordal differentiation. Here, we defined the somatic driver landscape of 104 cases of sporadic chordoma. We reveal somatic duplications of the notochordal transcription factor brachyury (T) in up to 27% of cases. These variants recapitulate the rearrangement architecture of the pathogenic germline duplications of T that underlie familial chordoma. In addition, we find potentially clinically actionable PI3K signalling mutations in 16% of cases. Intriguingly, one of the most frequently altered genes, mutated exclusively by inactivating mutation, was LYST (10%), which may represent a novel cancer gene in chordoma.Chordoma is a rare often incurable malignant bone tumour. Here, the authors investigate driver mutations of sporadic chordoma in 104 cases, revealing duplications in notochordal transcription factor brachyury (T), PI3K signalling mutations, and mutations in LYST, a potential novel cancer gene in chordoma.


Asunto(s)
Neoplasias Óseas/genética , Cordoma/genética , Proteínas Fetales/genética , Mutación , Proteínas de Dominio T Box/genética , Proteínas de Transporte Vesicular/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase Ia , Duplicación de Gen , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Polimorfismo de Nucleótido Simple
4.
Oncotarget ; 7(43): 69173-69187, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27732951

RESUMEN

Glioblastoma multiforme (GBM) carries a poor prognosis and continues to lack effective treatments. Glioblastoma stem cells (GSCs) drive tumor formation, invasion, and drug resistance and, as such, are the focus of studies to identify new therapies for disease control. Here, we identify the involvement of IKK and NF-κB signaling in the maintenance of GSCs. Inhibition of this pathway impairs self-renewal as analyzed in tumorsphere formation and GBM expansion as analyzed in brain slice culture. Interestingly, both the canonical and non-canonical branches of the NF-κB pathway are shown to contribute to this phenotype. One source of NF-κB activation in GBM involves the TGF-ß/TAK1 signaling axis. Together, our results demonstrate a role for the NF-κB pathway in GSCs and provide a mechanistic basis for its potential as a therapeutic target in glioblastoma.


Asunto(s)
Autorrenovación de las Células , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Quinasa I-kappa B/genética , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/genética , Interferencia de ARN , Ratas , Esferoides Celulares/metabolismo , Técnicas de Cultivo de Tejidos , Factor de Crecimiento Transformador beta/metabolismo
5.
Genes Cancer ; 5(1-2): 41-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24955217

RESUMEN

Activating mutations in KRAS are prevalent in cancer, but therapies targeted to oncogenic RAS have been ineffective to date. These results argue that targeting downstream effectors of RAS will be an alternative route for blocking RAS-driven oncogenic pathways. We and others have shown that oncogenic RAS activates the NF-κB transcription factor pathway and that KRAS-induced lung tumorigenesis is suppressed by expression of a degradation-resistant form of the IκBα inhibitor or by genetic deletion of IKKß or the RELA/p65 subunit of NF-κB. Here, genetic and pharmacological approaches were utilized to inactivate IKK in human primary lung epithelial cells transformed by KRAS, as well as KRAS mutant lung cancer cell lines. Administration of the highly specific IKKß inhibitor Compound A (CmpdA) led to NF-κB inhibition in different KRAS mutant lung cells and siRNA-mediated knockdown of IKKα or IKKß reduced activity of the NF-κB canonical pathway. Next, we determined that both IKKα and IKKß contribute to oncogenic properties of KRAS mutant lung cells, particularly when p53 activity is disrupted. Based on these results, CmpdA was tested for potential therapeutic intervention in the Kras-induced lung cancer mouse model (LSL-Kras (G12D)) combined with loss of p53 (LSL-Kras (G12D)/p53 (fl/fl)). CmpdA treatment was well tolerated and mice treated with this IKKß inhibitor presented smaller and lower grade tumors than mice treated with placebo. Additionally, IKKß inhibition reduced inflammation and angiogenesis. These results support the concept of targeting IKK as a therapeutic approach for oncogenic RAS-driven tumors with altered p53 activity.

6.
Mol Cell ; 45(6): 719-30, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22342344

RESUMEN

The IκB kinase (IKK) pathway is an essential mediator of inflammatory, oncogenic, and cell stress pathways. Recently IKK was shown to be essential for autophagy induction in mammalian cells independent of its ability to regulate NF-κB, but the mechanism by which this occurs is unclear. Here we demonstrate that the p85 regulatory subunit of PI3K is an IKK substrate, phosphorylated at S690 in vitro and in vivo in response to cellular starvation. Cells expressing p85 S690A or inhibited for IKK activity exhibit increased Akt activity following cell starvation, demonstrating that p85 phosphorylation is required for starvation-induced PI3K feedback inhibition. S690 is in a conserved region of the p85 cSH2 domain, and IKK-mediated phosphorylation of this site results in decreased affinity for tyrosine-phosphorylated proteins and decreased PI3K membrane localization. Finally, leucine deprivation is shown to be necessary and sufficient for starvation-induced, IKK-mediated p85 phosphorylation and PI3K feedback inhibition.


Asunto(s)
Quinasa I-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Inanición/metabolismo , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Línea Celular , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Secuencia Conservada , Retroalimentación Fisiológica , Fibroblastos/metabolismo , Humanos , Quinasa I-kappa B/genética , Leucina/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Fosfotirosina/metabolismo , Dominios Homologos src
7.
PLoS One ; 5(2): e9428, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20195534

RESUMEN

BACKGROUND: Activation of the transcription factor NF-kappaB by cytokines is rapid, mediated through the activation of the IKK complex with subsequent phosphorylation and degradation of the inhibitory IkappaB proteins. The IKK complex is comprised of two catalytic subunits, IKKalpha and IKKbeta, and a regulatory protein known as NEMO. Using cells from mice that are genetically deficient in IKKbeta or IKKalpha, or using a kinase inactive mutant of IKKbeta, it has been proposed that IKKbeta is critical for TNF-induced IkappaB phosphorylation/degradation through the canonical pathway while IKKalpha has been shown to be involved in the non-canonical pathway for NF-kappaB activation. These conclusions have led to a focus on development of IKKbeta inhibitors for potential use in inflammatory disorders and cancer. METHODOLOGY: Analysis of NF-kappaB activation in response to TNF in MEFs reveals that IKKbeta is essential for efficient phosphorylation and subsequent degradation of IkappaB alpha, yet IKKalpha contributes to the NF-kappaB activation response in these cells as measured via DNA binding assays. In HeLa cells, both IKKalpha and IKKbeta contribute to IkappaB alpha phosphorylation and NF-kappaB activation. A kinase inactive mutant of IKKbeta, which has been used as evidence for the critical importance of IKKbeta in TNF-induced signaling, blocks activation of NF-kappaB induced by IKKalpha, even in cells that are deficient in IKKbeta. CONCLUSIONS: These results demonstrate the importance of IKKalpha in canonical NF-kappaB activation, downstream of cytokine treatment of cells. The experiments suggest that IKKalpha will be a therapeutic target in inflammatory disorders.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Sondas de ADN/genética , Sondas de ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HeLa , Humanos , Quinasa I-kappa B/genética , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Interferencia de ARN
8.
Mol Cell Biol ; 28(16): 5061-70, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18541671

RESUMEN

Glutamate is a critical neurotransmitter of the central nervous system (CNS) and also an important regulator of cell survival and proliferation. The binding of glutamate to metabotropic glutamate receptors induces signal transduction cascades that lead to gene-specific transcription. The transcription factor NF-kappaB, which regulates cell proliferation and survival, is activated by glutamate; however, the glutamate receptor-induced signaling pathways that lead to this activation are not clearly defined. Here we investigate the glutamate-induced activation of NF-kappaB in glial cells of the CNS, including primary astrocytes. We show that glutamate induces phosphorylation, nuclear accumulation, DNA binding, and transcriptional activation function of glial p65. The glutamate-induced activation of NF-kappaB requires calcium-dependent IkappaB kinase alpha (IKKalpha) and IKKbeta activation and induces p65-IkappaBalpha dissociation in the absence of IkappaBalpha phosphorylation or degradation. Moreover, glutamate-induced IKK preferentially targets the phosphorylation of p65 but not IkappaBalpha. Finally, we show that the ability of glutamate to activate NF-kappaB requires cross-coupled signaling with the epidermal growth factor receptor. Our results provide insight into a glutamate-induced regulatory pathway distinct from that described for cytokine-induced NF-kappaB activation and have important implications with regard to both normal glial cell physiology and pathogenesis.


Asunto(s)
Receptores ErbB/metabolismo , Receptores de Glutamato/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Ácido Glutámico/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Ratones , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Fenilacetatos/farmacología , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
9.
Genes Dev ; 22(11): 1490-500, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519641

RESUMEN

While NF-kappaB is considered to play key roles in the development and progression of many cancers, the mechanisms whereby this transcription factor is activated in cancer are poorly understood. A key oncoprotein in a variety of cancers is the serine-threonine kinase Akt, which can be activated by mutations in PI3K, by loss of expression/activity of PTEN, or through signaling induced by growth factors and their receptors. A key effector of Akt-induced signaling is the regulatory protein mTOR (mammalian target of rapamycin). We show here that mTOR downstream from Akt controls NF-kappaB activity in PTEN-null/inactive prostate cancer cells via interaction with and stimulation of IKK. The mTOR-associated protein Raptor is required for the ability of Akt to induce NF-kappaB activity. Correspondingly, the mTOR inhibitor rapamycin is shown to suppress IKK activity in PTEN-deficient prostate cancer cells through a mechanism that may involve dissociation of Raptor from mTOR. The results provide insight into the effects of Akt/mTOR-dependent signaling on gene expression and into the therapeutic action of rapamycin.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Adaptadoras Transductoras de Señales , Regulación de la Expresión Génica , Células HeLa , Humanos , Masculino , Neoplasias de la Próstata , Proteínas/metabolismo , Proteína Reguladora Asociada a mTOR , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Células Tumorales Cultivadas
10.
J Immunol ; 178(8): 4803-10, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17404261

RESUMEN

Autoimmunity results from a breakdown in tolerance mechanisms that regulate autoreactive lymphocytes. We recently showed that during innate immune responses, secretion of IL-6 by dendritic cells (DCs) maintained autoreactive B cells in an unresponsive state. In this study, we describe that TLR4-activated DCs from lupus-prone mice are defective in repressing autoantibody secretion, coincident with diminished IL-6 secretion. Reduced secretion of IL-6 by MRL/lpr DCs reflected diminished synthesis and failure to sustain IL-6 mRNA production. This occurred coincident with lack of NF-kappaB and AP-1 DNA binding and failure to sustain IkappaBalpha phosphorylation. Analysis of individual mice showed that some animals partially repressed Ig secretion despite reduced levels of IL-6. This suggests that in addition to IL-6, DCs secrete other soluble factor(s) that regulate autoreactive B cells. Collectively, the data show that MRL/lpr mice are defective in DC/IL-6-mediated tolerance, but that some individuals maintain the ability to repress autoantibody secretion by an alternative mechanism.


Asunto(s)
Células Dendríticas/fisiología , Inmunoglobulinas/biosíntesis , Interleucina-6/fisiología , Lupus Eritematoso Sistémico/inmunología , Animales , Autoanticuerpos/biosíntesis , ADN/metabolismo , Tolerancia Inmunológica , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Transgénicos , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos B/fisiología , Receptor Toll-Like 4/fisiología
11.
J Biol Chem ; 281(18): 12521-5, 2006 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-16517600

RESUMEN

Phosphorylation of histone H3 protein at serine 10 is an important step in chromatin remodeling during transcriptional transactivation. IkappaB kinase-alpha (IKK-alpha) and Mitogen- and Stress-activated protein Kinases 1 and 2 (MSK1/2) have been shown to play key roles in the transcriptional regulation of immediate early genes such as c-fos. Interestingly, IKK-alpha and MSK1/2 have also been implicated as histone H3-Ser10 kinases. In this work, we have shown that MSK1/2 are required for epidermal growth factor (EGF)-induced, but not tumor necrosis factor-induced, histone H3-Ser10 phosphorylation, both globally and at specific promoters. Consistent with this, MSK1/2 are required for optimal immediate early c-fos transcription in response to EGF potentially through control of both H3-Ser10 and promoter-associated cAMP-response element-binding protein phosphorylation. Furthermore, MSK1/2 control EGF-induced IkappaB alpha promoter H3-Ser10 phosphorylation in the absence of elevated transcription. These studies demonstrate the existence of pathway-specific mechanisms to control histone H3-Ser10 phosphorylation and gene expression.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Histonas/química , Proteínas Quinasas S6 Ribosómicas 90-kDa/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Proteínas I-kappa B/metabolismo , Ratones , Inhibidor NF-kappaB alfa , Fosforilación , Regiones Promotoras Genéticas , Serina/química
12.
Genes Dev ; 20(2): 225-35, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16384933

RESUMEN

While Bcl-3 expression in cancer was originally thought to be limited to B-cell lymphomas with a 14;19 chromosomal translocation, more recent evidence indicates that expression of this presumptive oncoprotein is significantly more widespread in cancer. However, an oncogenic role for Bcl-3 has not been clearly identified. Experiments presented here indicate that Bcl-3 is inducible by DNA damage and is required for the induction of Hdm2 gene expression and the suppression of persistent p53 activity. Furthermore, constitutive expression of Bcl-3 suppresses DNA damage-induced p53 activation and inhibits p53-induced apoptosis through a mechanism that is at least partly dependent on the up-regulation of Hdm2. The results provide insight into a mechanism whereby altered expression of Bcl-3 leads to tumorigenic potential. Since Bcl-3 is required for germinal center formation, these results suggest functional similarities with the unrelated Bcl-6 oncoprotein in suppressing potential p53-dependent cell cycle arrest and apoptosis in response to somatic hypermutation and class switch recombination.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas del Linfoma 3 de Células B , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Mutación de Línea Germinal , Centro Germinal/metabolismo , Ratones , FN-kappa B/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Interferente Pequeño/metabolismo , Recombinación Genética , Factores de Transcripción , Células Tumorales Cultivadas , Regulación hacia Arriba
13.
Mol Cell Biol ; 25(19): 8444-55, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16166627

RESUMEN

Loss of glycogen synthase kinase 3beta (GSK-3beta) in mice results in embryonic lethality via hepatocyte apoptosis. Consistent with this result, cells from these mice have diminished nuclear factor kappaB (NF-kappaB) activity, implying a functional role for GSK-3beta in regulating NF-kappaB. Here, we have explored mechanisms by which GSK-3beta may control NF-kappaB function. We show that cytokine-induced IkappaB kinase activity and subsequent phosphorylation of IkappaBalpha, p105, and p65 are not affected by the absence of GSK-3beta activity. Furthermore, nuclear accumulation of p65 following tumor necrosis factor treatment is unaffected by the loss of GSK-3beta. However, NF-kappaB DNA binding activity is reduced in GSK-3beta null cells and in cells treated with a pharmacological inhibitor of GSK-3. Expression of certain NF-kappaB-regulated genes, such as IkappaBalpha and macrophage inflammatory protein 2, is minimally affected by the absence of GSK-3beta. Conversely, we have identified a subset of NF-kappaB-regulated genes, including those for interleukin-6 and monocyte chemoattractant protein 1, that require GSK-3beta for efficient expression. We show that efficient localization of p65 to the promoter regions of the interleukin-6 and monocyte chemoattractant protein 1 genes following tumor necrosis factor alpha treatment requires GSK-3beta. Therefore, GSK-3beta has profound effects on transcription in a gene-specific manner through a mechanism involving control of promoter-specific recruitment of NF-kappaB.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , FN-kappa B/metabolismo , Transcripción Genética , Animales , Apoptosis , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CXCL2 , Quimiocinas/metabolismo , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , ADN/química , ADN/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta , Inmunoprecipitación , Interleucina-6/metabolismo , Intestinos/citología , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
14.
Genes Dev ; 18(23): 2905-15, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15545623

RESUMEN

The c-Jun NH(2)-terminal kinase (JNK) has been implicated in both cell death and survival responses to different stimuli. Here we reexamine the function of JNK in tumor necrosis factor (TNF)-stimulated cell death using fibroblasts isolated from wild-type, Mkk4(-/-) Mkk7(-/-), and Jnk1(-/-) Jnk2(-/-) mice. We demonstrate that JNK can act to suppress TNF-stimulated apoptosis. However, we find that JNK can also potentiate TNF-stimulated necrosis by increasing the production of reactive oxygen species (ROS). Together, these data indicate that JNK can shift the balance of TNF-stimulated cell death from apoptosis to necrosis. Increased necrosis may represent a contributing factor in stress-induced inflammatory responses mediated by JNK.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa/fisiología , Animales , Muerte Celular/fisiología , MAP Quinasa Quinasa 4 , Ratones
15.
J Biol Chem ; 279(30): 31183-9, 2004 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-15155743

RESUMEN

Mitogenic activation of expression of immediate-early genes, such as c-fos, is controlled through signal-induced phosphorylation of constitutively bound transcription factors that is correlated with a nucleosomal response that involves inducible chromatin modifications, such as histone phosphorylation and acetylation. Here we have explored a potential role for the transcription factor NF-kappaB and its associated signaling components in mediating induction of c-fos gene expression downstream of epidermal growth factor (EGF)-dependent signaling. Here we show that EGF treatment of quiescent fibroblast does not induce the classical pathway of NF-kappaB activation through IkappaB kinase (IKK)-directed IkappaBalpha phosphorylation. Interestingly, efficient induction of c-fos transcription requires IKKalpha, one of the subunits of the IkappaB kinase complex. The NF-kappaB subunit, p65/RelA, is found constitutively associated with the c-fos promoter, and knock-out of this transcription factor significantly reduces c-fos gene expression. Importantly, EGF induces the recruitment of IKKalpha to the c-fos promoter to regulate promoter-specific histone H3 Ser(10) phosphorylation in a manner that is independent of p65/RelA. Collectively, our data demonstrate that IKKalpha and p65/RelA contribute significantly to EGF-induced c-fos gene expression in a manner independent of the classical, IkappaBalpha degradation, p65/RelA nuclear accumulation response pathway.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Genes fos , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN/genética , Expresión Génica/genética , Histonas/química , Histonas/metabolismo , Quinasa I-kappa B , Proteínas I-kappa B/metabolismo , Ratones , Ratones Noqueados , Inhibidor NF-kappaB alfa , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA
16.
Genes Dev ; 17(19): 2368-73, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14522944

RESUMEN

Cytokines, such as tumor necrosis factor-alpha (TNFalpha), potently inhibit the differentiation of mesenchymal cells and down-regulate the expression of Sox9 and MyoD, transcription factors required for chondrocyte and myocyte development. Previously, we demonstrated that NF-kappaB controls TNFalpha-mediated suppression of myogenesis through a mechanism involving MyoD mRNA down-regulation. Here, we show that NF-kappaB also suppresses chondrogenesis and destabilizes Sox9 mRNA levels. Multiple copies of an mRNA cis-regulatory motif (5'-ACUACAG-3') are necessary and sufficient for NF-kappaB-mediated Sox9 and MyoD down-regulation. Thus, in response to cytokine signaling, NF-kappaB modulates the differentiation of mesenchymal-derived cell lineages via RNA sequence-dependent, posttranscriptional down-regulation of key developmental regulators.


Asunto(s)
Diferenciación Celular/fisiología , Mesodermo/citología , FN-kappa B/metabolismo , Interferencia de ARN , Animales , Secuencia de Bases , Células Cultivadas , Condrocitos/citología , Condrocitos/fisiología , Secuencia Conservada , Regulación hacia Abajo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Mesodermo/metabolismo , Ratones , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , FN-kappa B/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Factor de Transcripción SOX9 , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Nature ; 423(6940): 659-63, 2003 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-12789343

RESUMEN

NF-kappaB is a principal transcriptional regulator of diverse cytokine-mediated processes and is tightly controlled by the IkappaB kinase complex (IKK-alpha/beta/gamma). IKK-beta and IKK-gamma are critical for cytokine-induced NF-kappaB function, whereas IKK-alpha is thought to be involved in other regulatory pathways. However, recent data suggest a role for IKK-alpha in NF-kappaB-dependent gene expression in response to cytokine treatment. Here we demonstrate nuclear accumulation of IKK-alpha after cytokine exposure, suggesting a nuclear function for this protein. Consistent with this, chromatin immunoprecipitation (ChIP) assays reveal that IKK-alpha was recruited to the promoter regions of NF-kappaB-regulated genes on stimulation with tumour-necrosis factor-alpha. Notably, NF-kappaB-regulated gene expression is suppressed by the loss of IKK-alpha and this correlates with a complete loss of gene-specific phosphorylation of histone H3 on serine 10, a modification previously associated with positive gene expression. Furthermore, we show that IKK-alpha can directly phosphorylate histone H3 in vitro, suggesting a new substrate for this kinase. We propose that IKK-alpha is an essential regulator of NF-kappaB-dependent gene expression through control of promoter-associated histone phosphorylation after cytokine exposure. These findings provide additional insight into the role of the IKK complex in NF-kappaB-regulated gene expression.


Asunto(s)
Regulación de la Expresión Génica , FN-kappa B/metabolismo , Nucleosomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Fibroblastos , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Quinasa I-kappa B , Proteínas I-kappa B/genética , Interleucina-6/genética , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/química , FN-kappa B/genética , Nucleosomas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa/farmacología
18.
J Biol Chem ; 278(5): 2963-8, 2003 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-12433922

RESUMEN

The transcription factor NF-kappa B has been shown to be predominantly cytoplasmically localized in the absence of an inductive signal. Stimulation of cells with inflammatory cytokines such as tumor necrosis factor alpha or interleukin-1 induces the degradation of I kappa B, the inhibitor of NF-kappa B, allowing nuclear accumulation of NF-kappa B and regulation of specific gene expression. The degradation of I kappa B is controlled initially by phosphorylation induced by the I kappa B kinase, which leads to ubiquitination and subsequent proteolysis of the inhibitor by the proteasome. We report here that NF-kappa B and I kappa B alpha (but not I kappa B beta) are also localized in the mitochondria. Stimulation of cells with tumor necrosis factor alpha leads to the phosphorylation of mitochondrial I kappa B alpha and its subsequent degradation by a nonproteasome-dependent pathway. Interestingly, expression of the mitochondrially encoded cytochrome c oxidase III and cytochrome b mRNAs were reduced by cytokine treatment of cells. Inhibition of activation of mitochondrial NF-kappa B by expression of the superrepressor form of I kappa B alpha inhibited the loss of expression of both cytochrome c oxidase III and cytochrome b mRNA. These data indicate that the NF-kappa B regulatory pathway exists in mitochondria and that NF-kappa B can negatively regulate mitochondrial mRNA expression.


Asunto(s)
Proteínas I-kappa B/análisis , Mitocondrias Hepáticas/ultraestructura , Mitocondrias/ultraestructura , FN-kappa B/análisis , FN-kappa B/genética , Animales , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas I-kappa B/genética , Inmunohistoquímica , Microscopía Electrónica , Mitocondrias/metabolismo , Mitocondrias Hepáticas/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , ARN Mensajero/genética , Ratas , Factor de Necrosis Tumoral alfa/farmacología , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA