Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 687627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220843

RESUMEN

Oral mucositis (OM) is a treatment-limiting adverse side effect of radiation and chemotherapy. Approximately 80% of patients undergoing radiotherapy (RT) for head and neck cancers (HNC) develop OM, representing a major unmet medical condition. Our understanding of the immunopathogenesis of OM is limited, due in part to the surprising paucity of information regarding healing mechanisms in the oral mucosa. RNAseq of oral tissue in a murine model that closely mimics human OM, showed elevated expression of IL-17 and related immune pathways in response to head and neck irradiation (HNI). Strikingly, mice lacking the IL-17 receptor (IL-17RA) exhibited markedly more severe OM. Restoration of the oral mucosa was compromised in Il17ra-/- mice and components associated with healing, including matrix metalloproteinase 3, 10 and IL-24 were diminished. IL-17 is typically associated with recruitment of neutrophils to mucosal sites following oral infections. Unexpectedly, in OM the absence of IL-17RA resulted in excessive neutrophil recruitment and immunopathology. Instead, neutrophil activation was IL-1R-driven in Il17ra-/- mice. Blockade of IL-1R and depletion of neutrophils lessened the severity of damage in these mice. Overall, we show IL-17 is protective in OM through multiple mechanisms including restoration of the damaged epithelia and control of the neutrophil response. We also present a clinically relevant murine model of human OM to improve mechanistic understanding and develop rational translational therapeutics.


Asunto(s)
Interleucina-17/metabolismo , Traumatismos por Radiación/metabolismo , Receptores de Interleucina-17/metabolismo , Estomatitis/metabolismo , Lengua/metabolismo , Cicatrización de Heridas , Animales , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Interleucina-1/metabolismo , Interleucina-17/genética , Ratones Noqueados , Infiltración Neutrófila , Traumatismos por Radiación/genética , Traumatismos por Radiación/inmunología , Traumatismos por Radiación/patología , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-17/genética , Transducción de Señal , Estomatitis/genética , Estomatitis/inmunología , Estomatitis/patología , Lengua/inmunología , Lengua/patología , Transcriptoma
2.
Biochem Pharmacol ; 183: 114349, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33245902

RESUMEN

Serotonin neurotransmission is largely governed by the regulation of the serotonin transporter (SERT). SERT is modulated in part by cholesterol, but the role of cholesterol and lipid signaling intermediates in regulating SERT are unknown. Serotonergic neurons were treated with statins to decrease cholesterol and lipid signaling intermediates. Contrary to reported decreases in 5-HT uptake after cholesterol depletion, biochemical and imaging methods both showed that statins increased 5-HT uptake in a fluoxetine-dependent manner. Simvastatin lowered the Km without changing Vmax for 5-HT or SERT distribution to the plasma membrane. Cholesterol repletion did not block enhanced 5-HT uptake by simvastatin but the enhanced uptake was blocked by lipid isoprenylation intermediates farnesyl pyrophosphate and geranylgeranyl pyrophosphate. Blockade of geranylgeranylation alone without statins also enhanced 5-HT uptake. Overall, this study revealed a specific neuronal effect of statin drugs and identified lipid signaling through geranylgeranylation within the isoprenylation pathway regulates SERT in a cholesterol-independent manner.


Asunto(s)
Colesterol/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Transducción de Señal/fisiología , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Células HEK293 , Humanos , Lípidos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología
3.
Sci Rep ; 9(1): 6688, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040372

RESUMEN

One third of newly diagnosed breast cancers in the US are early-stage lesions. The etiological understanding and treatment of these lesions have become major clinical challenges. Because breast cancer risk factors are often linked to aberrant nitric oxide (NO) production, we hypothesized that abnormal NO levels might contribute to the formation of early-stage breast lesions. We recently reported that the basal level of NO in the normal breast epithelia plays crucial roles in tissue homeostasis, whereas its reduction contributes to the malignant phenotype of cancer cells. Here, we show that the basal level of NO in breast cells plummets during cancer progression due to reduction of the NO synthase cofactor, BH4, under oxidative stress. Importantly, pharmacological deprivation of NO in prepubertal to pubertal animals stiffens the extracellular matrix and induces precancerous lesions in the mammary tissues. These lesions overexpress a fibrogenic cytokine, TGFß, and an oncogene, ERBB2, accompanied by the occurrence of senescence and stem cell-like phenotype. Consistently, normalization of NO levels in precancerous and cancerous breast cells downmodulates TGFß and ERBB2 and ameliorates their proliferative phenotype. This study sheds new light on the etiological basis of precancerous breast lesions and their potential prevention by manipulating the basal NO level.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Óxido Nítrico/biosíntesis , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Receptor ErbB-2/genética , Factor de Crecimiento Transformador beta/genética , Animales , Biomarcadores , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Expresión Génica , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Lesiones Precancerosas/patología , Receptor ErbB-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
Cancer Chemother Pharmacol ; 83(2): 237-254, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30426158

RESUMEN

PURPOSE: 3-(6-Methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (6-MOMIPP) is a novel indole-based chalcone that disrupts microtubules. The present study aims to define the mechanism through which 6-MOMIPP induces cell death and to evaluate the efficacy of the compound in penetrating the blood-brain barrier and inhibiting growth of glioblastoma xenografts. METHODS: The effects of 6-MOMIPP were evaluated in cultured U251 glioblastoma cells, using viability, flow cytometry, and tubulin polymerization assays. Scintillation proximity and tubulin crosslinking methods were used to identify the binding site of 6-MOMIPP on tubulin, and western blots were performed to define the signaling pathways that contribute to cell death. LC/MS assays were used to study the pharmacokinetic behavior of 6-MOMIPP in mice. Subcutaneous and intracerebral xenograft models were utilized to assess the effects of 6-MOMIPP on growth of U251 glioblastoma in vivo. RESULTS: The findings indicate that 6-MOMIPP targets the colchicine site on ß-tubulin. At concentrations ≥ 250 nm, 6-MOMIPP induces mitotic arrest, caspase activation and loss of cell viability. Cells are protected by caspase inhibitors, pointing to an apoptotic mechanism of cell death. Loss of cell viability is preceded by activation of Cdk1(Cdc2) and phosphorylation of Bcl-2 and Bcl-xL. Inhibition of both events with a Cdk1 inhibitor prevents cell death. 6-MOMIPP has broad activity against the viability of multiple glioblastoma, melanoma and lung carcinoma cell lines. Viability of normal cells, including differentiated neurons, is not significantly affected at a drug concentration (1 µM) that reduces viability in most cancer lines. Pharmacokinetic studies in mice show that concentrations of 6-MOMIPP in the brain mirror those in the plasma, indicating that 6-MOMIPP readily penetrates the blood-brain barrier. Studies with mice bearing human U251 glioblastoma xenografts demonstrate that 6-MOMIPP is effective in suppressing growth of subcutaneous and intracerebral tumors without causing general toxicity. CONCLUSIONS: The results indicate that 6-MOMIPP is a novel microtubule disruptor that targets the colchicine binding site on ß-tubulin to induce mitotic arrest and cell death. The ability of 6-MOMIPP to penetrate the blood-brain barrier and inhibit growth of glioblastoma xenografts suggests that it warrants further preclinical evaluation as potential small-molecule therapeutic that may have advantages in treating primary and metastatic brain tumors.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Indoles/farmacología , Mitosis , Piridinas/farmacología , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Indoles/farmacocinética , Ratones , Ratones Desnudos , Piridinas/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Gut Microbes ; 9(5): 400-421, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29469650

RESUMEN

BACKGROUND: Conditions of excess androgen in women, such as polycystic ovary syndrome (PCOS), often exhibit intergenerational transmission. One way in which the risk for PCOS may be increased in daughters of affected women is through exposure to elevated androgens in utero. Hyperandrogenemic conditions have serious health consequences, including increased risk for hypertension and cardiovascular disease. Recently, gut dysbiosis has been found to induce hypertension in rats, such that blood pressure can be normalized through fecal microbial transplant. Therefore, we hypothesized that the hypertension seen in PCOS has early origins in gut dysbiosis caused by in utero exposure to excess androgen. We investigated this hypothesis with a model of prenatal androgen (PNA) exposure and maternal hyperandrogenemia by single-injection of testosterone cypionate or sesame oil vehicle (VEH) to pregnant dams in late gestation. We then completed a gut microbiota and cardiometabolic profile of the adult female offspring. RESULTS: The metabolic assessment revealed that adult PNA rats had increased body weight and increased mRNA expression of adipokines: adipocyte binding protein 2, adiponectin, and leptin in inguinal white adipose tissue. Radiotelemetry analysis revealed hypertension with decreased heart rate in PNA animals. The fecal microbiota profile of PNA animals contained higher relative abundance of bacteria associated with steroid hormone synthesis, Nocardiaceae and Clostridiaceae, and lower abundance of Akkermansia, Bacteroides, Lactobacillus, Clostridium. The PNA animals also had an increased relative abundance of bacteria associated with biosynthesis and elongation of unsaturated short chain fatty acids (SCFAs). CONCLUSIONS: We found that prenatal exposure to excess androgen negatively impacted cardiovascular function by increasing systolic and diastolic blood pressure and decreasing heart rate. Prenatal androgen was also associated with gut microbial dysbiosis and altered abundance of bacteria involved in metabolite production of short chain fatty acids. These results suggest that early-life exposure to hyperandrogenemia in daughters of women with PCOS may lead to long-term alterations in gut microbiota and cardiometabolic function.


Asunto(s)
Andrógenos/efectos adversos , Disbiosis/microbiología , Hipertensión/etiología , Exposición Materna/efectos adversos , Síndrome del Ovario Poliquístico/complicaciones , Efectos Tardíos de la Exposición Prenatal/microbiología , Testosterona/análogos & derivados , Adipoquinas/metabolismo , Tejido Adiposo Blanco/metabolismo , Adulto , Andrógenos/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Presión Sanguínea , Disbiosis/etiología , Disbiosis/metabolismo , Disbiosis/fisiopatología , Ácidos Grasos Volátiles/metabolismo , Femenino , Microbioma Gastrointestinal , Frecuencia Cardíaca , Humanos , Hipertensión/metabolismo , Hipertensión/microbiología , Hipertensión/fisiopatología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Wistar , Testosterona/administración & dosificación , Testosterona/efectos adversos
6.
Brain Res Bull ; 91: 52-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23318273

RESUMEN

Recent studies have demonstrated that a preconditioning regimen (i.e., repeated low doses) of MDMA provides protection against the reductions in tissue concentrations of 5-HT and 5-HT transporter (SERT) density and/or expression produced by a subsequent binge regimen of MDMA. In the present study, the effects of preconditioning and binge treatment regimens of MDMA on SERT function were assessed by synaptosomal 5-HT uptake. Synaptosomal 5-HT uptake was reduced by 72% 7 days following the binge regimen (10 mg/kg, i.p. every 2 h for a total of 4 injections). In rats exposed to the preconditioning regimen of MDMA (daily treatment with 10 mg/kg for 4 days), the reduction in synaptosomal 5-HT uptake induced by a subsequent binge regimen was significantly less. Treatment with the preconditioning regimen alone resulted in a transient 46% reduction in 5-HT uptake that was evident 1 day, but not 7 days, following the last injection of MDMA. Furthermore, the preconditioning regimen of MDMA did not alter tissue concentrations of 5-HT, whereas the binge regimen of MDMA resulted in a long-term reduction of 40% of tissue 5-HT concentrations. The distribution of SERT immunoreactivity (ir) in membrane and endosomal fractions of the hippocampus also was evaluated following the preconditioning regimen of MDMA. There was no significant difference in the relative distribution of SERTir between these two compartments in control and preconditioned rats. The results demonstrate that SERT function is transiently reduced in response to a preconditioning regimen of MDMA, while long-term reductions in SERT function occur in response to a binge regimen of MDMA. Moreover, a preconditioning regimen of MDMA provides protection against the long-term reductions in SERT function evoked by a subsequent binge regimen of the drug. It is tempting to speculate that the neuroprotective effect of MDMA preconditioning results from a transient down-regulation in SERT function.


Asunto(s)
Química Encefálica/efectos de los fármacos , Alucinógenos/administración & dosificación , N-Metil-3,4-metilenodioxianfetamina/administración & dosificación , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Western Blotting , Química Encefálica/fisiología , Cromatografía Líquida de Alta Presión , Masculino , Microdiálisis/métodos , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Sinaptosomas/metabolismo
7.
Cancer Res ; 69(15): 6307-14, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19638578

RESUMEN

Alkylation chemotherapy has been a long-standing treatment protocol for human neoplasia. N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) is a direct-acting monofunctional alkylator. Temozolomide is a clinical chemotherapeutic equivalent requiring metabolic breakdown to the alkylating agent. Both chemicals have similar mechanistic efficacy against DNA mismatch repair-proficient tumor cells that lack expression of methylguanine methyltransferase. Clinically relevant concentrations of both agents affect replicating cells only after the first cell cycle. This phenomenon has been attributed to replication fork arrest at unrepaired O(6)-methyldeoxyguanine lesions mispaired with thymine during the first replication cycle. Here, we show, by several different approaches, that MNNG-treated tumor cells do not arrest within the second cell cycle. Instead, the population slowly traverses through mitosis without cytokinesis into a third cell cycle. The peak of both ssDNA and dsDNA breaks occurs at the height of the long mitotic phase. The majority of the population emerges from mitosis as multinucleated cells that subsequently undergo cell death. However, a very small proportion of cells, <1:45,000, survive to form new colonies. Taken together, these results indicate that multinucleation within the third cell cycle, rather than replication fork arrest within the second cell cycle, is the primary trigger for cell death. Importantly, multinucleation and cell death are consistently avoided by a small percentage of the population that continues to divide. This information should prove clinically relevant for the future design of enhanced cancer chemotherapeutics.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Ciclo Celular/efectos de los fármacos , Metilnitronitrosoguanidina/farmacología , Proteína Quinasa CDC2 , Ciclo Celular/genética , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Reparación de la Incompatibilidad de ADN , Metilasas de Modificación del ADN/deficiencia , Enzimas Reparadoras del ADN/deficiencia , Relación Dosis-Respuesta a Droga , Células HeLa , Histonas/metabolismo , Humanos , Fosforilación , Proteínas Supresoras de Tumor/deficiencia
8.
DNA Repair (Amst) ; 7(6): 951-69, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18468964

RESUMEN

Treatment with low concentrations of monofunctional alkylating agents induces a G2 arrest only after the second round of DNA synthesis in mammalian cells and requires a proficient mismatch repair (MMR) pathway. Here, we have investigated rapid alkylation-induced recruitment of DNA repair proteins to chromosomal DNA within synchronized populations of MMR proficient cells (HeLa MR) after N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) treatment. Within the first hour, the concentrations of MutS alpha and PCNA increase well beyond their constitutive chromosomally bound levels and MutL alpha is newly recruited to the chromatin-bound MutS alpha. Remarkably, immunoprecipitation experiments demonstrate rapid association of these proteins on the alkylation-damaged chromatin, even when DNA replication is completely blocked. The extent of association of PCNA and MMR proteins on the chromatin is dependent upon the concentration of MNNG and on the specific type of replication block. A subpopulation of the MutS alpha-associated PCNA also becomes monoubiquitinated, a known requirement for PCNA to interact with translesion synthesis (TLS) polymerases. In addition, chromatin-bound SMC1 and NBS1 proteins, associated with DNA double-strand-breaks (DSBs), become phosphorylated within 1-2h of exposure to MNNG. However, these activated proteins are not co-localized on the chromatin with MutS alpha in response to MNNG exposure. PCNA, MutS alpha/MutL alpha and activated SMC1/NBS1 remain chromatin-bound for at least 6-8h after alkylation damage. Thus, cells that are exposed to low levels of alkylation treatment undergo rapid recruitment to and/or activation of key proteins already on the chromatin without the requirement for DNA replication, apparently via different DNA-damage signaling pathways.


Asunto(s)
Disparidad de Par Base , Cromatina/metabolismo , Reparación del ADN , Metilnitronitrosoguanidina/farmacología , Western Blotting , Ciclo Celular , Inmunoprecipitación de Cromatina , Daño del ADN , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos
9.
Exp Cell Res ; 313(2): 292-304, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17157834

RESUMEN

The DNA mismatch repair (MMR) pathway contributes to the fidelity of DNA synthesis and recombination by correcting mispaired nucleotides and insertion/deletion loops (IDLs). We have investigated whether MMR protein expression, activity, and subcellular location are altered during discrete phases of the cell cycle in mammalian cells. Two distinct methods have been used to demonstrate that although physiological MMR protein expression, mismatch binding, and nick-directed MMR activity within the nucleus are at highest levels during S phase, MMR is active throughout the cell cycle. Despite equal MMR nuclear protein concentrations in S and G(2) phases, mismatch binding and repair activities within G(2) are significantly lower, indicating a post-translational decrease in MMR activity specific to G(2). We further demonstrate that typical co-localization of MutSalpha to late S phase replication foci can be disrupted by 2 microM N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). This concentration of MNNG does not decrease ongoing DNA synthesis nor induce cell cycle arrest until the second cell cycle, with long-term colony survival decreased by only 24%. These results suggest that low level alkylation damage can selectively disrupt MMR proofreading activity during DNA synthesis and potentially increase mutation frequency within surviving cells.


Asunto(s)
Ciclo Celular/genética , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/metabolismo , Animales , Núcleo Celular/química , Células Cultivadas , Replicación del ADN , Proteínas de Unión al ADN/análisis , Humanos , Antígeno Nuclear de Célula en Proliferación/análisis , Antígeno Nuclear de Célula en Proliferación/metabolismo
10.
BMC Mol Biol ; 6: 6, 2005 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-15766387

RESUMEN

BACKGROUND: The current investigation was undertaken to determine key steps differentiating G:T and G:A repair at the H-ras oncogenic hot spot within the nuclear environment because of the large difference in repair efficiency of these two mismatches. RESULTS: Electrophoretic mobility shift (gel shift) experiments demonstrate that DNA containing mismatched bases are recognized and bound equally efficiently by hMutSalpha in both MMR proficient and MMR deficient (hMLH1-/-) nuclear extracts. Competition experiments demonstrate that while hMutSalpha predictably binds the G:T mismatch to a much greater extent than G:A, hMutSalpha demonstrates a surprisingly equal ratio of competitive inhibition for both G:T and G:A mismatch binding reactions at the H-ras hot spot of mutation. Further, mismatch repair assays reveal almost 2-fold higher efficiency of overall G:A repair (5'-nick directed correct MMR to G:C and incorrect repair to T:A), as compared to G:T overall repair. Conversely, correct MMR of G:T --> G:C is significantly higher (96%) than that of G:A --> G:C (60%). CONCLUSION: Combined, these results suggest that initiation of correct MMR requires the contribution of two separate steps; initial recognition by hMutSalpha followed by subsequent binding. The 'avidity' of the binding step determines the extent of MMR pathway activation, or the activation of a different cellular pathway. Thus, initial recognition by hMutSalpha in combination with subsequent decreased binding to the G:A mismatch (as compared to G:T) may contribute to the observed increased frequency of incorrect repair of G:A, resulting in the predominant GGC --> GTC (Gly --> Val) ras-activating mutation found in a high percentage of human tumors.


Asunto(s)
Disparidad de Par Base/genética , Codón/genética , Reparación del ADN , Proteínas/genética , Proteínas ras/genética , Línea Celular Tumoral , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética/métodos , Células HCT116 , Humanos , Modelos Biológicos , Plásmidos/genética , Mutación Puntual/genética , Proto-Oncogenes/genética
11.
Glia ; 45(3): 238-48, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14730697

RESUMEN

Heregulin, a polypeptide growth factor, and forskolin, an adenylyl cyclase activator, synergistically stimulate expression of cyclin D3 and cell division in Schwann cells. Heregulin induces expression in Schwann cells of a luciferase reporter gene linked to the cyclin D3 promoter. Forskolin markedly augments reporter expression in the presence of heregulin. Deletion analysis identified several promoter sites that contribute to high-level reporter expression in heregulin- and forskolin-treated Schwann cells. A promoter fragment that contains 103 bp of 5'-flanking sequence produced significant reporter expression in heregulin- and forskolin-stimulated cells. Deletion of a consensus CCAAT site within this promoter fragment caused a nearly complete loss of reporter expression. Similar results were obtained when CCAAT site mutations were introduced into the promoter. Heregulin and forskolin increased steady-state levels of CCAAT/enhancer binding protein-beta (C/EBPbeta) in Schwann cells. Mobility shift assays identified proteins in Schwann cell nuclear extracts that formed stable complexes with the cyclin D3 CCAAT promoter element and were disrupted by anti-C/EBPbeta antibody. Transfection of Schwann cells with C/EBPbeta cDNA increased cyclin D3 reporter expression. In contrast to these results, mutation of a cAMP response element in the cyclin D3 promoter had only a modest effect on heregulin- and forskolin-stimulated reporter expression. These findings demonstrate that C/EBPbeta plays a key role in the heregulin and cAMP-dependent regulation of cyclin D3 expression in Schwann cells.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Colforsina/farmacología , Ciclinas/biosíntesis , Neurregulina-1/farmacología , Regiones Promotoras Genéticas/fisiología , Células de Schwann/efectos de los fármacos , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Ciclina D3 , Ciclinas/genética , Ciclinas/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Células de Schwann/metabolismo
12.
Mol Cancer Ther ; 1(9): 679-86, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12479364

RESUMEN

The signaling pathway for DNA damaging drug-triggered apoptosis was examined in a chemosensitive human neuroblastoma cell line, SH-SY5Y. Doxorubicin and etoposide induce rapid and extensive apoptosis in SH-SY5Y cells. After the drug treatment, p53 protein levels increase in the nucleus, leading to the induction of its transcription targets p21(Waf1/Cip1) and MDM2. Inactivation of p53, either by the human papillomavirus type 16 E6 protein or by a dominant-negative mutant p53 (R175H), completely protects SH-SY5Y cells from drug-triggered apoptosis. Cytochrome c and caspase-9 function downstream of p53 in mediating the drug-triggered apoptosis in SH-SY5Y cells. In drug-treated cells, cytochrome c is released, and caspase-9 becomes activated. Inactivation of p53 blocks cytochrome c release and caspase-9 activation. Furthermore, drug-induced cell death can be prevented by expression of a dominant-negative mutant of caspase-9. These findings define a molecular pathway for mediating DNA damaging drug-induced apoptosis in the human neuroblastoma SH-SY5Y cells and suggest that inactivation of essential components of this apoptotic pathway may confer drug resistance on neuroblastoma cells.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Genes p53 , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Caspasa 9 , Núcleo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Grupo Citocromo c/metabolismo , Daño del ADN , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Activación Enzimática , Etopósido/farmacología , Humanos , Immunoblotting , Microscopía Fluorescente , Mitocondrias/enzimología , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Células Tumorales Cultivadas
13.
Nat Cell Biol ; 4(11): 888-93, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12389034

RESUMEN

Nuclear factor-kappa B (NF-kappa B) promotes cell survival by upregulating expression of anti-apoptotic genes, a process that is antagonized by inhibitors of kappa B (I kappa B) factors. The only NF-kappa B family member known to be mutated in human cancer is NF-kappa B2 p100 (ref. 2), a factor with I kappa B activity. Here, we report the isolation from irradiated mouse tumour cells of a complex that induces caspase-8 activity in cell-free assays and identify p100 as an essential component of this complex. Expression of p100 profoundly sensitizes cells to death-receptor-mediated apoptosis through a pathway that is independent of I kappa B-like activity. The carboxyl terminus of p100 contains a death domain that is absent from all known tumour-derived mutants. This death domain mediates recruitment of p100 into death machinery complexes after ligand stimulation and is essential for p100's pro-apoptotic activity. p100 also sensitizes NIH3T3 cells to apoptosis triggered by oncogenic Ras, resulting in a marked inhibition of transformation that is rescued by suppression of endogenous caspase-8. These observations thus identify an I kappa B-independent apoptotic activity of NF-kappa B2 p100 and help explain its unique tumour suppressor role.


Asunto(s)
Apoptosis , FN-kappa B/fisiología , Animales , Caspasa 8 , Caspasas/metabolismo , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Sistema Libre de Células , Colorimetría , Cicloheximida/farmacología , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Humanos , Immunoblotting , Espectrometría de Masas , Ratones , Mutación , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B , Células 3T3 NIH , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Retroviridae/genética , Factores de Tiempo , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...