Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 112(6): 1042-51, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25742482

RESUMEN

BACKGROUND: Activation of the PI3K/mTOR and Hedgehog (Hh) signalling pathways occurs frequently in biliary tract cancer (BTC). Crosstalk between these pathways occurs in other gastrointestinal cancers. The respective signalling inhibitors rapamycin and vismodegib may inhibit BTC synergistically and suppress cancer stem cells (CSCs). METHODS: Gene expression profiling for p70S6k and Gli1 was performed with BTC cell lines. Tumour and pathway inhibitory effects of rapamycin and vismodegib were investigated in BTC preclinical models and CSCs. RESULTS: Rapamycin and vismodegib synergistically reduced BTC cell viability and proliferation. This drug combination arrested BTC Mz-ChA-1 cells in the G1 phase but had no significant effect on the cell cycle of BTC Sk-ChA-1 cells. Combined treatment inhibited the proliferation of CSCs and ALDH-positive cells. Nanog and Oct-4 expression in CSCs was decreased by the combination treatment. Western blotting results showed the p-p70S6K, p-Gli1, p-mTOR, and p-AKT protein expression were inhibited by the combination treatment in BTC cells. In an Mz-ChA-1 xenograft model, combination treatment resulted in 80% inhibition of tumour growth and prolonged tumour doubling time. In 4 of 10 human BTC specimens, tumour p-p70S6K and Gli1 protein expression levels were decreased with the combination treatment. CONCLUSIONS: Targeted inhibition of the PI3K/mTOR and Hhpathways indicates a new avenue for BTC treatment with combination therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Sistema Biliar/tratamiento farmacológico , Proteínas Hedgehog/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Anilidas/administración & dosificación , Animales , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/metabolismo , Neoplasias del Sistema Biliar/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fase G1/efectos de los fármacos , Fase G1/genética , Perfilación de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Desnudos , Proteína Homeótica Nanog , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Piridinas/administración & dosificación , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína con Dedos de Zinc GLI1
2.
J Hosp Infect ; 139: 175-191, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37419189

RESUMEN

OBJECTIVE: This systematic review and network meta-analysis (NMA) comprehensively compared the effectiveness of different mouth rinses in reducing the viral load/infectivity of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) (Part I), alleviating clinical symptoms or severity of disease (Part II), and decreasing the incidence of SARS-CoV-2 infection (Part III). METHODS: Randomized controlled trials (RCTs) and non-randomized controlled trials (NRCTs) with restrictions were searched up to 3rd March 2023. Twenty-three studies (22 RCTs and one NRCT) met the inclusion criteria for this systematic review. RESULTS: Five RCTs (454 patients and nine interventions) in Part I were eligible for NMA. The NMA results showed that, in comparison with no rinse, sodium chloride (NaCl) was the most effective mouth rinse for reducing the viral load, followed by povidone-iodine (PVP-I), ß-cyclodextrin + citrox (CDCM), hydrogen peroxide (HP), chlorhexidine gluconate (CHX), cetylpyridinium chloride (CPC), placebo and hypochlorous acid (HClO). However, these results were not significant. Based on surface under the cumulative ranking curve scores, PVP-I was likely to be the most efficacious mouth rinse for reducing SARS-CoV-2 viral load, followed by CDCM, HP, NaCl, CHX, CPC, placebo, no rinse and HClO. CONCLUSION: Due to heterogeneity of the primary studies, the effectiveness of different mouth rinses to reduce viral infectivity, improve clinical symptoms or prevent SARS-CoV-2 infection remains inconclusive.


Asunto(s)
COVID-19 , Humanos , Antisépticos Bucales/uso terapéutico , Povidona Yodada , SARS-CoV-2 , Cloruro de Sodio/uso terapéutico , Metaanálisis en Red , Peróxido de Hidrógeno , Boca
3.
Br J Cancer ; 106(2): 243-7, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22187039

RESUMEN

Accumulated evidence shows that EZH2 is deregulated in a wide range of cancer types, and it has a crucial role in stem cell maintenance and tumour development. Therefore, blocking EZH2 expression or activity may represent a promising strategy for anticancer treatment. In this review, we address the current understanding of the mechanisms underlying EZH2 regulation alongside the function of EZH2 gene targets that are involved in cancer progression. Finally, we will describe cancer therapies that target EZH2 or its downstream cascades, which could potentially reverse the oncogenic and stemness properties of the tumour cells to suppress cancer progression and recurrence.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neoplasias/fisiopatología , Factores de Transcripción/fisiología , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/patología , Complejo Represivo Polycomb 2 , Factores de Transcripción/metabolismo
4.
Nat Cell Biol ; 3(3): 245-52, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11231573

RESUMEN

Amplification or overexpression of HER-2/neu in cancer cells confers resistance to apoptosis and promotes cell growth. The cellular localization of p21Cip1/WAF1 has been proposed to be critical either in promoting cell survival or in inhibiting cell growth. Here we show that HER-2/neu-mediated cell growth requires the activation of Akt, which associates with p21Cip1/WAF1 and phosphorylates it at threonine 145, resulting in cytoplasmic localization of p21Cip1/WAF1. Furthermore, blocking the Akt pathway with a dominant-negative Akt mutant restores the nuclear localization and cell-growth-inhibiting activity of p21Cip1/WAF1. Our results indicate that HER-2/neu induces cytoplasmic localization of p21Cip1/WAF1 through activation of Akt to promote cell growth, which may have implications for the oncogenic activity of HER-2/neu and Akt.


Asunto(s)
División Celular/fisiología , Ciclinas/metabolismo , Citoplasma/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Células 3T3 , Adenocarcinoma/genética , Adenocarcinoma/patología , Secuencias de Aminoácidos , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular/genética , Fraccionamiento Celular , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Femenino , Fibroblastos , Regulación de la Expresión Génica , Ratones , Microscopía Fluorescente , Fosforilación , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Receptor ErbB-2/genética , Transducción de Señal/fisiología , Transfección
5.
Nat Cell Biol ; 3(11): 973-82, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11715018

RESUMEN

HER-2/neu amplification or overexpression can make cancer cells resistant to apoptosis and promotes their growth. p53 is crucial in regulating cell growth and apoptosis, and is often mutated or deleted in many types of tumour. Moreover, many tumours with a wild-type gene for p53 do not have normal p53 function, suggesting that some oncogenic signals suppress the function of p53. In this study, we show that HER-2/neu-mediated resistance to DNA-damaging agents requires the activation of Akt, which enhances MDM2-mediated ubiquitination and degradation of p53. Akt physically associates with MDM2 and phosphorylates it at Ser166 and Ser186. Phosphorylation of MDM2 enhances its nuclear localization and its interaction with p300, and inhibits its interaction with p19ARF, thus increasing p53 degradation. Our study indicates that blocking the Akt pathway mediated by HER-2/neu would increase the cytotoxic effect of DNA-damaging drugs in tumour cells with wild-type p53.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinas/metabolismo , Células 3T3 , Animales , Línea Celular Transformada , Núcleo Celular/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Daño del ADN , Proteína p300 Asociada a E1A , Etopósido/farmacología , Humanos , Ratones , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-mdm2 , Receptor ErbB-2/genética , Transactivadores/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo
6.
Nat Cell Biol ; 3(9): 802-8, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533659

RESUMEN

Epidermal growth factor receptor (EGFR) has been detected in the nucleus in many tissues and cell lines. However, the potential functions of nuclear EGFR have largely been overlooked. Here we demonstrate that nuclear EGFR is strongly correlated with highly proliferating activities of tissues. When EGFR was fused to the GAL4 DNA-binding domain, we found that the carboxy terminus of EGFR contained a strong transactivation domain. Moreover, the receptor complex bound and activated AT-rich consensus-sequence-dependent transcription, including the consensus site in cyclin D1 promoter. By using chromatin immunoprecipitation assays, we further demonstrated that nuclear EGFR associated with promoter region of cyclin D1 in vivo. EGFR might therefore function as a transcription factor to activate genes required for highly proliferating activities.


Asunto(s)
Membrana Celular/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/citología , Células Epiteliales/fisiología , Receptores ErbB/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Mama , Línea Celular , Cromatina/fisiología , Secuencia de Consenso , Receptores ErbB/genética , Femenino , Humanos , Cinética , Ratones , Datos de Secuencia Molecular , Embarazo , Transporte de Proteínas , Proteínas Recombinantes/metabolismo , Transfección , Células Tumorales Cultivadas , Útero/citología , Útero/metabolismo
7.
Nat Med ; 6(2): 189-95, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10655108

RESUMEN

Because HER-2/neu overexpression is important in cancer development, we looked for a method of suppressing the cell transformation mediated by HER-2/neu overexpression. We have identified that the DNA-binding protein PEA3, which is encoded by a previously isolated gene of the ets family, specifically targeted a DNA sequence on the HER-2/neu promoter and downregulated the promoter activity. Expression of PEA3 resulted in preferential inhibition of cell growth and tumor development of HER-2/neu-overexpressing cancer cells. This is a new approach to targeting HER-2/neu overexpression and also provides a rationale to the design for repressors of diseases caused by overexpression of pathogenic genes.


Asunto(s)
Transformación Celular Neoplásica , Regulación de la Expresión Génica/fisiología , Receptor ErbB-2/genética , Factores de Transcripción/fisiología , Células 3T3 , Animales , Secuencia de Bases , División Celular/fisiología , Cartilla de ADN , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Neoplasias Experimentales/patología , Regiones Promotoras Genéticas , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo
8.
Oncogene ; 39(4): 946-949, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31537904

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Science ; 229(4717): 976-8, 1985 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-2992090

RESUMEN

The neu oncogene, identified in ethylnitrosourea-induced rat neuroglioblastomas, had strong homology with the erbB gene that encodes the epidermal growth factor receptor. This homology was limited to the region of erbB encoding the tyrosine kinase domain. It was concluded that the neu gene is a distinct novel gene, as it is not coamplified with sequences encoding the EGF receptor in the genome of the A431 tumor line and it maps to human chromosome 17.


Asunto(s)
Antígenos de Neoplasias/genética , ADN de Neoplasias/genética , Proteínas de Neoplasias/genética , Oncogenes , Receptores de Superficie Celular/genética , Animales , Mapeo Cromosómico , Cromosomas Humanos 16-18 , Receptores ErbB , Genes , Ligamiento Genético , Humanos , Neuroblastoma/genética , Neuroglía , Ratas
10.
Dis Esophagus ; 22(2): 127-32, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19021681

RESUMEN

Activated nuclear factor-kappa B (NF-kappaB) in the pretreatment cancer tissue of patients with localized esophageal adenocarcinoma (LEA) undergoing preoperative chemoradiation is associated with poor prognosis. It is known that constitutively activated NF-kappaB prior to any therapy portends poor prognosis, and it is also known that activated NF-kappaB in the treated specimen is associated with poor prognosis. However, the prognosis of patients who have treatment-induced activation of NF-kappaB (meaning their cancers activate NF-kappaB during or after therapy) is not been reported. We hypothesized that the treatment-induced activation of NF-kappaB would impart poor prognosis similar to that imparted by constitutively activated NF-kappaB cancer. Patients with LEA who had undergone preoperative chemoradiation plus surgery and had pre- and post-therapy cancer tissue available were selected. Pre- and post-therapy cancer tissues were stained by immunohistochemistry for nuclear staining of NF-kappaB. The overall survival (OS) and disease-free survival were assessed and compared for patients who had intrinsic constitutively activated NF-kappaB cancer with those who had induced activation of NF-kappaB only post-therapy. A total of 41 patients with LEA were investigated. Twenty-five patients had NF-kappaB positive cancer at baseline, and 16 had NF-kappaB negative cancer at baseline but became positive post-therapy. There was no difference in the location, histology grade, clinical stage, or the curative resection (RO) resection rate in the two populations. OS (P = 0.71), disease-free survival (P = 0.86), and median survivals (Converters: 24 months [95% confidence intervals: 7.78 to 40.22]vs. Nonconverters: 34.13 months [95% confidence intervals: 3.54 to 64.27]) were not different between the two groups. Our data suggest that activation of NF-kappaB in response to stress/injury of therapy leads to poor OS. These results need to be confirmed in a larger number of patients. It may be that only pre-therapy evaluation of NF-kappaB is insufficient to assess prognosis of patients with LEA. Additional implications include that when effective anti-NF-kappaB therapies become available, they may have to be considered in patients whose cancers do not have constitutively activated NF-kappaB or cancer may have to be monitored during therapy with biomarker assessments.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , FN-kappa B/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/cirugía , Adenocarcinoma/terapia , Terapia Combinada , Supervivencia sin Enfermedad , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/cirugía , Neoplasias Esofágicas/terapia , Esofagectomía , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Pronóstico
11.
Nanotechnology ; 19(44): 445714, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19081800

RESUMEN

In this study, we fabricated electrospun silica nanofiber membranes and investigated their use in biomolecular sensing. The diameter, porosity and surface-to-volume ratio of nanofiber membranes were investigated under different fabrication conditions. Using this type of nanofiber membrane, enzyme-linked immunosorbent assay (ELISA) was performed, and the results were compared with those obtained with conventional ELISA using polystyrene well plates. The minimum detectable concentration was determined as 0.19 ng ml(-1) (1.6 pM), which is 32 times lower than that of conventional ELISA. In addition, the detection time for all processes for the nanofiber membrane was reduced to 1 h, compared with 1 day for conventional ELISA. The increased sensitivity, faster reaction time, and affordability of the nanofiber membrane make it well suited for bio-chip use.

12.
Oncogene ; 37(2): 208-217, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28925391

RESUMEN

Inhibitors against poly (ADP-ribose) polymerase (PARP) are promising targeted agents currently used to treat BRCA-mutant ovarian cancer and are in clinical trials for other cancer types, including BRCA-mutant breast cancer. To enhance the clinical response to PARP inhibitors (PARPis), understanding the mechanisms underlying PARPi sensitivity is urgently needed. Here, we show enhancer of zeste homolog 2 (EZH2), an enzyme that catalyzes H3 lysine trimethylation and associates with oncogenic function, contributes to PARPi sensitivity in breast cancer cells. Mechanistically, upon oxidative stress or alkylating DNA damage, PARP1 interacts with and attaches poly-ADP-ribose (PAR) chains to EZH2. PARylation of EZH2 by PARP1 then induces PRC2 complex dissociation and EZH2 downregulation, which in turn reduces EZH2-mediated H3 trimethylation. In contrast, inhibition of PARP by PARPi attenuates alkylating DNA damage-induced EZH2 downregulation, thereby promoting EZH2-mediated gene silencing and cancer stem cell property compared with PARPi-untreated cells. Moreover, the addition of an EZH2 inhibitor sensitizes the BRCA-mutant breast cells to PARPi. Thus, these results may provide a rationale for combining PARP and EZH2 inhibition as a therapeutic strategy for BRCA-mutated breast and ovarian cancers.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , ADP-Ribosilación/efectos de los fármacos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Daño del ADN , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Silenciador del Gen , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Mol Cell Biol ; 12(6): 2739-48, 1992 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1350324

RESUMEN

In an attempt to study potential feedback regulation of the neu oncogene, we have found that the neu oncogene product specifically represses its own promoter activity. Deletion analysis indicated a 140-bp region (nucleotides -312 to -173 relative to the ATG initiation codon) in the rat neu promoter responsible for neu autorepression. Gel shift assays and methylation interference analysis further demonstrated that a GGTGGGGGGG sequence (nucleotides -243 to -234 relative to the ATG initiation codon) in this 140-bp region interacts with specific protein complexes. The GGTGGGGGGG sequence (GTG element), which functions as an enhancer, is sufficient to cause neu-mediated repression in a heterologous promoter. Furthermore, it produces different gel shift patterns with nuclear extracts from neu-transformed cell lines and their parental lines, suggesting that a transcriptional factor(s) interacting with this enhancer element has been perturbed by the introduction of neu. Taken together, the data presented in this report show that (i) the neu oncogene product autorepresses its own promoter, (ii) the neu promoter contains a novel enhancer, and (iii) neu autorepression is mediated through this enhancer, likely by inhibition of the enhancer activity.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Células Cultivadas , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Retroalimentación , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química , Regiones Promotoras Genéticas , ARN Mensajero/genética , Ratas , Receptor ErbB-2 , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética
14.
Mol Cell Biol ; 11(1): 354-62, 1991 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1670896

RESUMEN

Amplification or overexpression or both of either the c-myc or the human neu (C-erbB-2) gene are common events in many primary human tumors. Coamplification or overexpression or both of both genes have been reported in some breast cancers. The possibility of cooperation between the c-myc and the normal rat neu (c-neu) genes in transforming cells was examined. Surprisingly, the expression of c-myc in B104-1-1 cells, and activated rat neu oncogene (neu*)-transformed NIH 3T3 line, resulted in morphologic reversion. This reversion was found to be a consequence of a transcription-repressive action of c-myc on the neu gene via a 140-bp fragment on the neu gene promoter. The effective concentration of a positive factor(s) interacting with this fragment seemed to be lowered by the expression of c-myc. Our findings lend support to arguments concerning the long-suspected function of c-myc as a transcriptional modulator. They also imply that an oncogene such as c-myc, or possibly the rapidly explored class that encodes transcription factors, under certain conditions may act to reverse a transformed phenotype that is induced by another oncogene instead of contributing positively towards the transformation process. Therefore, the activity of an oncogene may depend on the environment in which it is expressed. In addition, we may have identified the neu gene as a cellular target gene of negative regulation by c-myc.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Oncogenes , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteínas Proto-Oncogénicas/genética , Animales , Secuencia de Bases , Sitios de Unión , Análisis Mutacional de ADN , Proteínas de Unión al ADN/fisiología , Amplificación de Genes , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Neoplásico/genética , Receptor ErbB-2 , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/fisiología , Transcripción Genética , Transfección
15.
Mol Cell Biol ; 11(4): 1875-82, 1991 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1672439

RESUMEN

We used chloramphenicol acetyltransferase (CAT) assays to identify and characterize cis-acting elements responsible for rat neu promoter function. Deletion of a region of the neu promoter (-504 to -312) resulted in a marked decrease in CAT activity, indicating that this promoter region corresponds to a positive cis-acting element. Using band shift assays and methylation interference analyses, we further identified a specific protein-binding sequence, AAGATAAAACC (-466 to -456), that binds a specific trans-acting factor termed RVF (for EcoRV factor on the neu promoter). The RVF-binding site is required for maximum transcriptional activity of the rat neu promoter. This same sequence is also found in the corresponding regions of both human and mouse neu promoters. Furthermore, this sequence can enhance the CAT activity driven by a minimum promoter of the thymidine kinase gene in an orientation-independent manner, and thus it behaves as an enhancer. Our results demonstrate that RVF is the major DNA-binding protein contributing to enhancer activity. In addition, Southwestern (DNA-protein) blot analysis using the RVF-binding site as a probe points to a 60-kDa polypeptide as a potential candidate for RVF.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Animales , Secuencia de Bases , Sitios de Unión , Unión Competitiva , Cloranfenicol O-Acetiltransferasa/metabolismo , Secuencia de Consenso , Expresión Génica , Humanos , Metilación , Datos de Secuencia Molecular , Ratas , Receptor ErbB-2 , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
16.
Mol Cell Biol ; 10(12): 6306-15, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2123292

RESUMEN

A 2.4-kb rat neu genomic DNA fragment that hybridized to the 5'-most coding sequence of the rat neu cDNA was cloned. S1 nuclease mapping identified multiple transcriptional initiation sites. DNA sequence analysis revealed that this fragment contained 64 bp of the first intron, 81 bp of the first exon, and the upstream noncoding sequence of the neu gene. The sequence immediately upstream of the translation start site was G + C rich (greater than 75%) and contained a consensus CCAAT sequence despite the absence of a TATA box. An Sp1-binding site was found, in addition to various sequence motifs common to the promoters of the human neu gene (erbB2), the epidermal growth factor receptor gene, and the simian virus 40 enhancer. A 2.2-kb EcoRI-Narl fragment containing sequences upstream from the 3'-most transcriptional start site was fused to the bacterial chloramphenicol acetyltransferase reporter gene and shown to promote transcription efficiently. A series of promoter deletion constructs was made, and results from transfection and subsequent chloramphenicol acetyltransferase assays suggested the presence of multiple cis-acting elements that contributed either positively or negatively to the transcription activity. Cotransfection competition experiments using subcloned cis-acting elements confirmed the existence of trans-acting factors interacting with these DNA fragments. In addition, a gel retardation assay was performed to demonstrate the physical binding of nuclear factors to certain fragments. The results complemented those of the deletion studies and led us to conclude that transcriptional regulation of the neu proto-oncogene involves at least one negative and three positive trans-acting factors interacting with different cis-acting elements along the neu gene promoter.


Asunto(s)
Regulación de la Expresión Génica , Oncogenes , Transcripción Genética , Animales , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Clonación Molecular , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Mapeo Restrictivo , Transfección , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
17.
Mol Cell Biol ; 11(3): 1745-50, 1991 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1671710

RESUMEN

The adenovirus type 5 early region 1A (E1A) gene was introduced into neu-transformed B104-1-1 cells. Cells that expressed E1A possessed reduced transforming activity in vitro and reduced tumorigenicity in nude mice. These results demonstrate that the E1A gene products can act negatively to suppress the transformed phenotype in neu-transformed cells.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Oncogénicas Virales/fisiología , Oncogenes , Proteínas Proto-Oncogénicas/genética , Proteínas Precoces de Adenovirus , Animales , Southern Blotting , División Celular , Línea Celular , Expresión Génica , Ratones , Ratones Desnudos , Neoplasias Experimentales/genética , Receptor ErbB-2 , Transfección
18.
Mol Cell Biol ; 19(12): 8075-82, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10567533

RESUMEN

The adenovirus type 5 early region 1A gene (E1A) has previously been known as an immortalization oncogene because E1A is required for transforming oncogenes, such as ras and E1B, to transform cells in primary cultures. However, E1A has also been shown to downregulate the overexpression of the Her-2/neu oncogene, resulting in suppression of transformation and tumorigenesis induced by that oncogene. In addition, E1A is able to promote apoptosis induced by anticancer drugs, irradiation, and serum deprivation. Many tyrosine kinases, such as the epidermal growth factor receptor, Her-2/Neu, Src, and Axl, are known to play a role in oncogenic signals in transformed cells. To study the mechanism underlying the E1A-mediated tumor-suppressing function, we exploited a modified tyrosine kinase profile assay (D. Robinson, F. Lee, T. Pretlow, and H.-J. Kung, Proc. Natl. Acad. Sci. USA 93:5958-5962, 1996) to identify potential tyrosine kinases regulated by E1A. Reverse transcription (RT)-PCR products were synthesized with two degenerate primers derived from the conserved motifs of various tyrosine kinases. A tyrosine kinase downregulated by E1A was identified by analyzing the AluI-digested RT-PCR products. We isolated the DNA fragment of interest and found that E1A negatively regulated the expression of the transforming receptor tyrosine kinase Axl at the transcriptional level. To study whether downregulation of the Axl receptor is involved in E1A-mediated growth suppression, we transfected axl cDNA into E1A-expressing cells (ip1-E1A) to establish cells that overexpressed Axl. The Axl ligand Gas6 triggered a greater mitogenic effect in these ip1-E1A-Axl cells than in ip1-E1A control cells and protected the Axl-expressing cells from serum deprivation-induced apoptosis. These results indicate that downregulation of the Axl receptor by E1A is involved in E1A-mediated growth suppression and E1A-induced apoptosis and thereby contributes to E1A's antitumor activities.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Apoptosis , Péptidos y Proteínas de Señalización Intercelular , Proteínas Oncogénicas/metabolismo , Proteínas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas E1A de Adenovirus/genética , División Celular , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Humanos , Mitógenos , Proteínas Oncogénicas/genética , Proteínas/genética , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/genética , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Tirosina Quinasa del Receptor Axl
19.
Mol Cell Biol ; 15(11): 6398-405, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7565792

RESUMEN

Human pro-tumor necrosis factor (pro-TNF) is a type II transmembrane protein with a highly conserved 76-residue leader sequence. We have analyzed the behavior, both in a microsomal translocational system and by transfection, of a series of mutants with deletions from the cytoplasmic, transmembrane, and linking domains. Cytoplasmic deletions included the Arg doublet at -49 and -48 and/or the Lys doublet at -58 and -57; additional mutants included deletion of residues -73 to -55 and -73 to -55, -49, and -48. The transmembrane and linking domain mutants included deletions in the -42 to -35 region, combined with the deletion of residues -32 to -1. Two hybrid mutants combined the cytoplasmic deletions with the deletion of residues -32 to -1. All of the cytoplasmic deletion mutants were properly translocated, as were the transmembrane deletion mutants with deletions up to residues -36, -35, -32 to -1, although the last one exhibited reduced efficiency; further incremental deletions, including deletions of residues -38 to -35 and -32 to -1, completely blocked translocation. Both hybrid mutants were effectively translocated; furthermore, transfection analysis revealed competent expression and maturation of both the cytoplasmic and hybrid mutants. Thus, proper expression and maturation of human pro-TNF can be accomplished with as few as approximately 12 of the 26 residues of the native transmembrane domain and with a net negative charge in the cytoplasmic domain flanking the transmembrane region.


Asunto(s)
Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Señales de Clasificación de Proteína/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Membrana Celular/metabolismo , Sistema Libre de Células , Células Cultivadas , Chlorocebus aethiops , Perros , Retículo Endoplásmico Rugoso/metabolismo , Humanos , Iones , Datos de Secuencia Molecular , ARN Mensajero/genética , Eliminación de Secuencia , Relación Estructura-Actividad
20.
Mol Cell Biol ; 20(2): 672-83, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10611246

RESUMEN

The neu (c-erbB-2) proto-oncogene encodes a tyrosine kinase receptor that is overexpressed in 20 to 30% of human breast tumors. Herein, cyclin D1 protein levels were increased in mammary tumors induced by overexpression of wild-type Neu or activating mutants of Neu in transgenic mice and in MCF7 cells overexpressing transforming Neu. Analyses of 12 Neu mutants in MCF7 cells indicated important roles for specific C-terminal autophosphorylation sites and the extracellular domain in cyclin D1 promoter activation. Induction of cyclin D1 by NeuT involved Ras, Rac, Rho, extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38, but not phosphatidylinositol 3-kinase. NeuT induction of the cyclin D1 promoter required the E2F and Sp1 DNA binding sites and was inhibited by dominant negative E2F-1 or DP-1. Neu-induced transformation was inhibited by a cyclin D1 antisense or dominant negative E2F-1 construct in Rat-1 cells. Growth of NeuT-transformed mammary adenocarcinoma cells in nude mice was blocked by the cyclin D1 antisense construct. These results demonstrate that E2F-1 mediates a Neu-signaling cascade to cyclin D1 and identify cyclin D1 as a critical downstream target of neu-induced transformation.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Transformación Celular Neoplásica/patología , Ciclina D1/metabolismo , Sistema de Señalización de MAP Quinasas , Receptor ErbB-2/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Ciclina D1/antagonistas & inhibidores , Ciclina D1/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Mutación/genética , Regiones Promotoras Genéticas/genética , Proto-Oncogenes Mas , ARN sin Sentido/genética , ARN sin Sentido/fisiología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3 , Factor de Transcripción DP1 , Activación Transcripcional , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA