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
2.
Nucleic Acids Res ; 47(2): 824-842, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30517763

RESUMEN

The downregulation of the DNA damage response (DDR) enables aggressive tumors to achieve uncontrolled proliferation against replication stress, but the mechanisms underlying this process in tumors are relatively complex. Here, we demonstrate a mechanism through which a distinct E3 ubiquitin ligase, ITCH, modulates DDR machinery in triple-negative breast cancer (TNBC). We found that expression of a nuclear form of ITCH was significantly increased in human TNBC cell lines and tumor specimens. Phosphorylation of ITCH at Ser257 by AKT led to the nuclear localization of ITCH and ubiquitination of H1.2. The ITCH-mediated polyubiquitination of H1.2 suppressed RNF8/RNF168-dependent formation of 53BP1 foci, which plays important roles in DDR. Consistent with these findings, impaired ITCH nuclear translocation and H1.2 polyubiquitination sensitized cells to replication stress and limited cell growth and migration. AKT activation of ITCH-H1.2 axis may confer TNBC cells with a DDR repression to counteract the replication stress and increase cancer cell survivorship and growth potential.


Asunto(s)
Neoplasias de la Mama/enzimología , Núcleo Celular/metabolismo , Daño del ADN , Histonas/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Transporte Activo de Núcleo Celular , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Replicación del ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/fisiología , Femenino , Células HEK293 , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/fisiología
3.
Mol Cell Biol ; 37(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28533220

RESUMEN

Chronic inflammation plays important roles in cancer initiation and progression. Resolving chronic inflammation or blocking inflammatory signal transduction may prevent cancer development. Here, we report that the combined low-dose use of two anti-inflammatory drugs, aspirin and triptolide, reduces spontaneous lung cancer incidence from 70% to 10% in a mouse model. Subsequent studies reveal that such treatment has little effect on resolving chronic inflammatory conditions in the lung, but it significantly blocks the NF-κB-mediated expression of proliferation and survival genes in cancer cells. Furthermore, triptolide and aspirin induce distinct mechanisms to potentiate each other to block NF-κB nuclear localization stimulated by inflammatory cytokines. While aspirin directly inhibits IκB kinases (IKKs) to phosphorylate IκBα for NF-κB activation, triptolide does not directly target IKKs or other factors that mediate IKK activation. Instead, it requires p53 to inhibit IκBα phosphorylation and degradation. Triptolide binds to and activates p38α and extracellular signal-regulated kinase 1/2 (ERK1/2), which phosphorylate and stabilize p53. Subsequently, p53 competes with IκBα for substrate binding to IKKß and thereby blocks IκBα phosphorylation and NF-κB nuclear translocation. Inhibition of p38α and ERK1/2 or p53 mutations could abolish the inhibitory effects of triptolide on NF-κB. Our study defines a new p53-dependent mechanism for blocking NF-κB survival pathways in cancer cells.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antiinflamatorios/uso terapéutico , Aspirina/uso terapéutico , Diterpenos/uso terapéutico , Inmunosupresores/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , FN-kappa B/inmunología , Fenantrenos/uso terapéutico , Proteína p53 Supresora de Tumor/inmunología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios no Esteroideos/administración & dosificación , Aspirina/administración & dosificación , Línea Celular Tumoral , Diterpenos/administración & dosificación , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/uso terapéutico , Femenino , Inmunosupresores/administración & dosificación , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Ratones , Fenantrenos/administración & dosificación , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Cell Rep ; 3(5): 1389-97, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23643536

RESUMEN

Chronic inflammation has a tight cause-and-effect relationship with DNA damage by inflicting tissue damage and increasing cancer risk. Rrm2b, a key enzyme in de novo deoxyribonucleotide synthesis, is involved in DNA damage repair, but its role in cancer development has yet to be demonstrated. In this work, Rrm2b gene loss led to severe numerical and structural chromosome abnormalities that caused ATM activation, inducing p-Ser85 IKKγ/NEMO and IκB kinase (IKK). NF-κB consequently induced by IKK triggered sustained IL-6 expression that constitutively activated STAT3 in Rrm2b-deficient cells. High plasma interleukin-6 (IL-6) and associated hematologic disorders were observed in Rrm2b-/- mice, and 30%-40% of aged Rrm2b heterozygous knockout mice developed plasma cell neoplasms and suffered from progressive splenomegaly and ascites. The genetic ablation of IL-6 suppressed STAT3 induction and delayed disease onset in Rrm2b-/- mice, extending their lifespan. Thus, Rrm2b plays a crucial role in maintaining chromosomal stability and preventing chronic-inflammation-associated tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Interleucina-6/metabolismo , Leucemia de Células Plasmáticas/metabolismo , Ribonucleótido Reductasas/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Transformación Celular Neoplásica , Quimiocina CCL2/metabolismo , ADN/metabolismo , Genotipo , Heterocigoto , Quinasa I-kappa B/metabolismo , Interleucina-6/genética , Cariotipificación , Leucemia de Células Plasmáticas/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Unión Proteica , Ribonucleótido Reductasas/deficiencia , Ribonucleótido Reductasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción ReIA/metabolismo
5.
Hepatology ; 57(5): 1919-30, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23175432

RESUMEN

UNLABELLED: Growing evidence indicates that deregulation of microRNAs (miRNAs) contributes to tumorigenesis. Dysregulation of miR-657 has been observed in several types of cancers, but its biological function is still largely unknown. Our results showed that miR-657 expression can be induced by hepatitis viral proteins and is significantly increased in hepatocellular carcinoma (HCC) tissues. Moreover, introduction of miR-657 dramatically increases proliferation and colony formation of HCC cells in vitro and induces tumor development in immunodeficient mice. Further studies showed that miR-657 directly targets the transducin-like enhancer protein 1 (TLE1) 3' untranslated region (UTR) and activates nuclear factor kappa B (NF-κB) pathways that contribute to hepatocarcinogenesis. CONCLUSION: This study identified a mechanism whereby miRNA-657 contributed to HCC through novel cancer pathways and provides new insights into the potential molecular mechanisms of hepatic carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica/metabolismo , Neoplasias Hepáticas/patología , MicroARNs/metabolismo , FN-kappa B/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Proteínas Co-Represoras , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Técnicas In Vitro , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Transfección , Trasplante Heterólogo
6.
Sci Rep ; 2: 822, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23139867

RESUMEN

RRM2B is the DNA damage-inducible small subunit of ribonucleotide reductase, the rate-limiting enzyme in de novo deoxyribonucleoside triphosphate synthesis. Although RRM2B is implicated in DNA repair and the maintenance of mitochondrial DNA content, the regulation and function of RRM2B in senescence have not been previously established. Here, we show that RRM2B is highly induced in a p53-dependent manner during senescence in primary human fibroblast IMR90 cells and is expressed at higher levels in senescent precancerous human prostatic intraepithelial neoplasm lesions compared to adjacent normal prostate glands. Paradoxically, silencing RRM2B expression leads to an increase in the level of reactive oxygen species, mitochondrial membrane depolarization, and premature senescence in a p38MAPK- and p53-dependent manner in young fibroblasts. Consistently, induction of senescence is accelerated in Rrm2b deficient mouse embryo fibroblasts. Our data demonstrate that RRM2B is induced by stress signals prior to the onset of senescence and prevents premature oxidative stress-induced senescence.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Senescencia Celular , Estrés Oxidativo , Ribonucleótido Reductasas/metabolismo , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular , Daño del ADN , Reparación del ADN , Humanos , Masculino , Ratones , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Mol Cancer Ther ; 10(2): 269-78, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21216934

RESUMEN

Deregulation of the expression of p53R2, a p53-inducible homologue of the R2 subunit of ribonucleotide reductase, has been found in various human cancer tissues; however, the roles p53R2 plays in cancer progression and malignancy remain controversial. In the present study, we examined changes in gene expression profiles associated with p53R2 in cancer cells, using the analysis of cDNA microarray. Gene set enrichment analysis identified that the gene set regulating cell-cycle progression was significantly enriched in p53R2-silencing human oropharyngeal carcinoma KB cells. Attenuation of p53R2 expression significantly reduced p21 expression and moderately increased cyclin D1 expression in both wild-type p53 cancer cells (KB and MCF-7) and mutant p53 cancer cells (PC3 and MDA-MB-231). Conversely, overexpression of p53R2-GFP resulted in an increase in the expression of p21 and decrease in the expression of cyclin D1, which correlated with reduced cell population in S-phase in vitro and suppressed growth in vivo. Furthermore, the MAP/ERK kinase inhibitor PD98059 partially abolished modulation of p21 and cyclin D1 expression by p53R2. Moreover, under the conditions of nonstress and adriamycin-induced genotoxic stress, attenuation of p53R2 in KB cells significantly increased phosphorylated H2AX, which indicates that attenuation of p53R2 may enhance DNA damage induced by adriamycin. Overall, our study shows that p53R2 may suppress cancer cell proliferation partially by upregulation of p21 and downregulation of cyclin D1; p53R2 plays critical roles not only in DNA damage repair but also in proliferation of cancer cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/genética , Neoplasias/genética , Neoplasias/metabolismo , Ribonucleótido Reductasas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Humanos , Células KB , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias/patología , Fosforilación , Ribonucleótido Reductasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Clin Oncol ; 29(10): e257-61, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21220613
9.
Proc Natl Acad Sci U S A ; 105(47): 18519-24, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19015526

RESUMEN

Ribonucleotide reductase small subunit p53R2 was identified as a p53 target gene that provides dNTP for DNA damage repair. However, the slow transcriptional induction of p53R2 in RNA may not be rapid enough for prompt DNA damage repair, which has to occur within a few hours of damage. Here, we demonstrate that p53R2 becomes rapidly phosphorylated at Ser(72) by ataxia telangiectasia mutated (ATM) within 30 min after genotoxic stress. p53R2, as well as its heterodimeric partner RRM1, are associated with ATM in vivo. Mutational studies further indicate that ATM-mediated Ser(72) phosphorylation is essential for maintaining p53R2 protein stability and conferring resistance to DNA damage. The mutation of Ser(72) on p53R2 to alanine results in the hyperubiquitination of p53R2 and reduces p53R2 stability. MDM2, a ubiquitin ligase for p53, interacts and facilitates ubiquitination of the S72A-p53R2 mutant more efficiently than WT-p53R2 after DNA damage in vivo. Our results strongly suggest a novel mechanism for the regulation of p53R2 activity via ATM-mediated phosphorylation at Ser(72) and MDM2-dependent turnover of p53R2 dephosphorylated at the same residue.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Daño del ADN , Proteínas de Unión al ADN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ribonucleótido Reductasas/metabolismo , Serina/metabolismo , Proteínas Supresoras de Tumor/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/química , Reparación del ADN , Humanos , Fosforilación , Ribonucleótido Reductasas/química , Rayos Ultravioleta
10.
Proc Natl Acad Sci U S A ; 103(28): 10741-6, 2006 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-16818881

RESUMEN

The c-Jun N-terminal kinases (JNKs) are key regulators of inflammation and interfere with insulin action in cultured cells and whole animals. Obesity increases total JNK activity, and JNK1, but not JNK2, deficiency results in reduced adiposity and improved insulin sensitivity. Interestingly, a higher-than-normal level of JNK activation is observed in Jnk2(-/-) mice, particularly in the liver, indicating an interaction between the isoforms that might have masked the metabolic activity of JNK2 in isolated mutant mice. To address the role of the JNK2 isoform in metabolic homeostasis, we intercrossed Jnk1(-/-) and Jnk2(-/-) mice and examined body weight and glucose metabolism in the resulting mutant allele combinations. Among all of the viable genotypes examined, we observed only reduced body weight and increased insulin sensitivity in Jnk1(-/-) and Jnk1(+/-)Jnk2(-/-) mice. These two groups of mice also exhibited reduced total JNK activity and cytokine expression in liver tissue compared with all other genotypes examined. These data indicate that the JNK2 isoform is also involved in metabolic regulation, but its function is not obvious when JNK1 is fully expressed because of regulatory crosstalk between the two isoforms.


Asunto(s)
Resistencia a la Insulina , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Proteína Quinasa 9 Activada por Mitógenos/fisiología , Obesidad/enzimología , Animales , Línea Celular , Marcadores Genéticos , Glucosa/metabolismo , Humanos , Resistencia a la Insulina/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiología , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/genética , Obesidad/genética
11.
Proc Natl Acad Sci U S A ; 103(28): 10544-51, 2006 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-16807293

RESUMEN

A major link between inflammation and cancer is provided by NF-kappaB transcription factors. Ikkbeta(Deltahep) mice, which specifically lack IkappaB kinase beta (IKKbeta), an activator of NF-kappaB, in hepatocytes, are unable to activate NF-kappaB in response to proinflammatory stimuli, such as TNF-alpha. Surprisingly, Ikkbeta(Deltahep) mice are hypersusceptible to diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Because defective NF-kappaB activation promotes sustained c-Jun N-terminal kinase (JNK) activation in cells exposed to TNF-alpha, whose expression is induced by DEN, and JNK activity is required for normal hepatocyte proliferation, we examined whether increased susceptibility to DEN-induced hepatocarcinogenesis in Ikkbeta(Deltahep) mice requires JNK activation. Hepatocytes express both JNK1 and JNK2, but previous studies indicate that JNK1 is more important for hepatocyte proliferation. We therefore investigated this hypothesis using mice homozygous for a JNK1 deficiency either in wild-type or Ikkbeta(Deltahep) backgrounds. In both cases, mice lacking JNK1 were much less susceptible to DEN-induced hepatocarcinogenesis. This impaired tumorigenesis correlated with decreased expression of cyclin D and vascular endothelial growth factor, diminished cell proliferation, and reduced tumor neovascularization. Whereas hepatocyte-specific deletion of IKKbeta augmented DEN-induced hepatocyte death and cytokine-driven compensatory proliferation, disruption of JNK1 abrogated this response. In addition to underscoring the importance of JNK1-mediated hepatocyte death and compensatory proliferation, these results strongly suggest that the control of tissue renewal through the IKK and JNK pathways plays a key role in liver carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/enzimología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Alquilantes/toxicidad , Animales , Carcinoma Hepatocelular/patología , Dietilnitrosamina/toxicidad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/genética
12.
Cell ; 124(3): 601-13, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16469705

RESUMEN

The proinflammatory cytokine tumor necrosis factor (TNF) alpha signals both cell survival and death. The biological outcome of TNFalpha treatment is determined by the balance between NF-kappaB and Jun kinase (JNK) signaling; NF-kappaB promotes survival, whereas JNK enhances cell death. Critically, identity of a JNK substrate that promotes TNFalpha-induced apoptosis has been outstanding. Here we show that TNFalpha-mediated JNK activation accelerates turnover of the NF-kappaB-induced antiapoptotic protein c-FLIP, an inhibitor of caspase-8. This is not due to direct c-FLIP phosphorylation but depends on JNK-mediated phosphorylation and activation of the E3 ubiquitin ligase Itch, which specifically ubiquitinates c-FLIP and induces its proteasomal degradation. JNK1 or Itch deficiency or treatment with a JNK inhibitor renders mice resistant in three distinct models of TNFalpha-induced acute liver failure, and cells from these mice do not display inducible c-FLIP(L) ubiquitination and degradation. Thus, JNK antagonizes NF-kappaB during TNFalpha signaling by promoting the proteasomal elimination of c-FLIP(L).


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Activación Enzimática , Femenino , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Fallo Hepático Agudo/etiología , Fallo Hepático Agudo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Modelos Biológicos , Embarazo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral , Transducción de Señal , Receptores Señuelo del Factor de Necrosis Tumoral
13.
Cell ; 120(5): 649-61, 2005 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-15766528

RESUMEN

TNFalpha is a pleiotropic cytokine that induces either cell proliferation or cell death. Inhibition of NF-kappaB activation increases susceptibility to TNFalpha-induced death, concurrent with sustained JNK activation, an important contributor to the death response. Sustained JNK activation in NF-kappaB-deficient cells was suggested to depend on reactive oxygen species (ROS), but how ROS affect JNK activation was unclear. We now show that TNFalpha-induced ROS, whose accumulation is suppressed by mitochondrial superoxide dismutase, cause oxidation and inhibition of JNK-inactivating phosphatases by converting their catalytic cysteine to sulfenic acid. This results in sustained JNK activation, which is required for cytochrome c release and caspase 3 cleavage, as well as necrotic cell death. Treatment of cells or experimental animals with an antioxidant prevents H(2)O(2) accumulation, JNK phosphatase oxidation, sustained JNK activity, and both forms of cell death. Antioxidant treatment also prevents TNFalpha-mediated fulminant liver failure without affecting liver regeneration.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Estrés Oxidativo/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antioxidantes/farmacología , Células COS , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Chlorocebus aethiops , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Células HeLa , Humanos , Peróxido de Hidrógeno/metabolismo , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/enzimología , Fallo Hepático Agudo/prevención & control , Ratones , Ratones Noqueados , Necrosis/enzimología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Ácidos Sulfénicos/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/farmacología , Factor de Necrosis Tumoral alfa/farmacología
14.
Mol Cell Biol ; 24(13): 5914-22, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15199146

RESUMEN

Oxidative stress and reactive oxygen species (ROS) can elicit and modulate various physiological and pathological processes, including cell death. However, the mechanisms controlling ROS-induced cell death are largely unknown. Data from this study suggest that receptor-interacting protein (RIP) and tumor necrosis factor receptor (TNFR)-associated factor 2 (TRAF2), two key effector molecules of TNF signaling, are essential for ROS-induced cell death. We found that RIP(-/-) or TRAF2(-/-) mouse embryonic fibroblasts (MEF) are resistant to ROS-induced cell death when compared to wild-type cells, and reconstitution of RIP and TRAF2 gene expression in their respective deficient MEF cells restored their sensitivity to H(2)O(2)-induced cell death. We also found that RIP and TRAF2 form a complex upon H(2)O(2) exposure, but without the participation of TNFR1. The colocalization of RIP with a membrane lipid raft marker revealed a possible role of lipid rafts in the transduction of cell death signal initiated by H(2)O(2). Finally, our results demonstrate that activation of c-Jun NH(2)-terminal kinase 1 is a critical event downstream of RIP and TRAF2 in mediating ROS-induced cell death. Therefore, our study uncovers a novel signaling pathway regulating oxidative stress-induced cell death.


Asunto(s)
Estrés Oxidativo , Proteínas/fisiología , Animales , Muerte Celular , Línea Celular , Humanos , Peróxido de Hidrógeno/farmacología , Células Jurkat , Ratones , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas/genética , Proteínas/metabolismo , Especies Reactivas de Oxígeno/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal , Factor 2 Asociado a Receptor de TNF , Transfección
15.
Immunity ; 19(5): 725-37, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14614859

RESUMEN

IkappaB kinase beta (IKKbeta) is required for NF-kappaB activation and suppression of TNFalpha-mediated liver apoptosis. To investigate how IKKbeta suppresses apoptosis, we generated hepatocyte-specific Ikkbeta knockout mice, Ikkbeta(Deltahep), which exhibit little residual NF- kappaB activity but are healthy with normal liver function. Unexpectedly, Ikkbeta(Deltahep) mice are slightly more sensitive than controls to LPS-induced liver apoptosis but are highly susceptible to liver destruction following concanavalin A (ConA)-induced T cell activation. Unlike LPS, a potent inducer of circulating TNFalpha, ConA exerts cytotoxic effects through cell-bound TNFalpha, which activates type 1 and 2 TNF receptors (TNFR). While TNFR2 does not contribute to NF-kappaB activation, it is important for ConA-induced JNK activation, which is augmented by the absence of IKKbeta. Using JNK-deficient mice we show that JNK is required for ConA-induced liver damage. Thus, the antiapoptotic function of IKKbeta, which is most critical in situations that involve cell-bound TNFalpha, is mediated partially through attenuation of JNK activity.


Asunto(s)
Apoptosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígenos CD/metabolismo , Concanavalina A/metabolismo , Quinasa I-kappa B , Proteínas Quinasas JNK Activadas por Mitógenos , Lipopolisacáridos/metabolismo , Hígado/patología , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral , Linfocitos T/fisiología
16.
Dev Cell ; 4(4): 521-33, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12689591

RESUMEN

Microtubules (MTs) play an important role in elaboration and maintenance of axonal and dendritic processes. MT dynamics are modulated by MT-associated proteins (MAPs), whose activities are regulated by protein phosphorylation. We found that a member of the c-Jun NH(2)-terminal protein kinase (JNK) subgroup of MAP kinases, JNK1, is involved in regulation of MT dynamics in neuronal cells. Jnk1(-/-) mice exhibit disrupted anterior commissure tract formation and a progressive loss of MTs within axons and dendrites. MAP2 and MAP1B polypeptides are hypophosphorylated in Jnk1(-/-) brains, resulting in compromised ability to bind MTs and promote their assembly. These results suggest that JNK1 is required for maintaining the cytoskeletal integrity of neuronal cells and is a critical regulator of MAP activity and MT assembly.


Asunto(s)
Diferenciación Celular/genética , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/crecimiento & desarrollo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Neuronas/enzimología , Animales , Axones/metabolismo , Axones/patología , Axones/ultraestructura , Sistema Nervioso Central/ultraestructura , Dendritas/metabolismo , Dendritas/patología , Dendritas/ultraestructura , Ratones , Ratones Noqueados , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Microtúbulos/ultraestructura , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/ultraestructura , Fosforilación , Unión Proteica/genética
17.
Nature ; 420(6913): 333-6, 2002 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-12447443

RESUMEN

Obesity is closely associated with insulin resistance and establishes the leading risk factor for type 2 diabetes mellitus, yet the molecular mechanisms of this association are poorly understood. The c-Jun amino-terminal kinases (JNKs) can interfere with insulin action in cultured cells and are activated by inflammatory cytokines and free fatty acids, molecules that have been implicated in the development of type 2 diabetes. Here we show that JNK activity is abnormally elevated in obesity. Furthermore, an absence of JNK1 results in decreased adiposity, significantly improved insulin sensitivity and enhanced insulin receptor signalling capacity in two different models of mouse obesity. Thus, JNK is a crucial mediator of obesity and insulin resistance and a potential target for therapeutics.


Asunto(s)
Resistencia a la Insulina/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Obesidad/enzimología , Obesidad/fisiopatología , Tejido Adiposo/enzimología , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Dieta , Modelos Animales de Enfermedad , Eliminación de Gen , Homeostasis , Hiperinsulinismo/complicaciones , Hiperinsulinismo/enzimología , Hiperinsulinismo/genética , Hiperinsulinismo/fisiopatología , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina/genética , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Ratones Obesos , Proteína Quinasa 8 Activada por Mitógenos , Proteína Quinasa 9 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Proteínas Quinasas Activadas por Mitógenos/genética , Obesidad/complicaciones , Obesidad/genética , Fosfoproteínas/metabolismo , Fosforilación , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Arthritis Rheum ; 46(3): 818-23, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11920420

RESUMEN

OBJECTIVE: Previous studies have demonstrated that inhibition of c-Jun N-terminal kinase (JNK) decreases joint destruction in the rat adjuvant arthritis model. The present study was undertaken to investigate whether selective loss of JNK-2 function decreases joint destruction in JNK-2 knockout mice, in order to determine the role of this isoform in inflammatory arthritis. METHODS: Passive collagen-induced arthritis (CIA) was induced in Jnk2(-/-) and wild-type mice by administering anti-type II collagen antibodies. Arthritis was assessed daily using a semiquantitative clinical scoring system. Fibroblast-like synoviocytes (FLS) were prepared from Jnk2(-/-) and wild-type mice, and JNK protein expression was determined by Western blot analysis. Matrix metalloproteinase 13 (MMP-13) expression was determined by Northern blot analysis, and activator protein 1 (AP-1) binding activity by electromobility shift assay (EMSA). RESULTS: The JNK protein level in Jnk2(-/-) mice with CIA was 22% of that in wild-type mice with CIA (P < 0.001), and mainly the 46-kd isoform was expressed in the former group. Surprisingly, clinical arthritis was slightly more severe in the Jnk2(-/-) mice. Histologic scores for synovial inflammation were not significantly different. However, Safranin O-stained sections from the Jnk2(-/-) mice exhibited significantly less joint damage. Although joint destruction was decreased in Jnk2(-/-) mice with CIA, EMSA and Northern blot analysis of total joint extracts revealed similar levels of AP-1 binding and MMP-13 expression in Jnk2(-/-) and wild-type mice. The lack of correlation with AP-1 activity and MMP expression was probably because non-FLS cells in the joint may express more JNK-1 than do FLS. CONCLUSION: JNK-2 is a determinant of matrix degradation, but it has little effect on inflammation in arthritis. Complete inhibition of MMP expression and joint destruction will likely require combined JNK-1 and JNK-2 inhibition.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Experimental/patología , Articulaciones/patología , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Animales , Colagenasas/genética , Expresión Génica , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Noqueados/genética , Proteína Quinasa 9 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...