Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
2.
Am J Surg ; 217(4): 794-799, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30638725

RESUMEN

BACKGROUND: The purpose of this study was to evaluate the websites of general surgery residency programs in the United States and Puerto Rico. METHODS: Electronic Residency Application Service (ERAS) websites (n = 254) were accessed between October 2016 and January 2017 and evaluated for content, including: education, resident and faculty information, program environment and specific recruitment incentives. RESULTS: General information, such as conference information, rotations, and faculty information were available for more than 80% of programs. However, specific details about residents, faculty, and applicant information were noticeably lacking. This included resident biographical data and research, faculty names and research endeavors, alumni locations and fellowship placement. Applicant information, specifically board score requirements, were present in less than half of websites nationally. Regionally, websites from the Midwest were the most detailed in the information they provided, while those from the Northeast were the least informative. CONCLUSIONS: As a primary information source for potential future residents, general surgery programs need to maximize the content and utility of their websites in order to attract prospective residents to their programs.


Asunto(s)
Educación de Postgrado en Medicina , Cirugía General/educación , Internet , Internado y Residencia , Humanos , Selección de Personal , Puerto Rico , Estados Unidos
3.
Cancer Res ; 65(10): 3986-92, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15899786

RESUMEN

Kinase suppressor of Ras1 (KSR1) interacts with several mitogen-activated protein (MAP) kinase pathway components, including Raf, MAP/extracellular signal-regulated kinase (ERK) kinase (MEK), and ERK, and acts as a positive regulator of the Ras signaling cascade. Previous studies have shown that exposure of cells to the anticancer agent cisplatin (cis-diamminedichloroplatinum, CDDP) is associated with changes in multiple signal transduction pathways, including c-Jun-NH2-kinase, ERK, and p38 pathways. Moreover, ERK activation has been linked to changes in cell survival following CDDP treatment. In this report, we have examined the effects of KSR1 expression on the sensitivity of cells to CDDP-induced apoptosis. Loss of KSR1 expression in mouse embryo fibroblasts (MEFs) derived from KSR1 knockout mice (KSR-/- MEF) is associated with decreased CDDP-induced ERK activation and increased resistance to CDDP-induced apoptosis compared with wild-type MEFs (KSR+/+ MEF). Furthermore, transduction of KSR-/- MEFs and MCF-7 breast cancer cells with wild-type KSR1 resulted in enhanced ERK activation following CDDP exposure and increased sensitivity to CDDP. In addition, inhibition of ERK activation by exposing MEFs to the MEK1/2-specific inhibitors PD98059 and U0126 protected both KSR+/+ and KSR-/- MEFs cells from CDDP-induced apoptosis. These results indicate that KSR1-mediated regulation of ERK activity represents a novel determinant of CDDP sensitivity of cancer cells.


Asunto(s)
Cisplatino/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética
4.
Surg Clin North Am ; 97(3): 641-656, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28501252

RESUMEN

Primary colorectal lymphoma, carcinoids (neuroendocrine tumors), and gastrointestinal stromal tumors comprise a small subset of all colorectal cancers. Their features are unique, and their treatment varies from that of colorectal adenocarcinoma. Appropriate identification is key in the management of these tumors.


Asunto(s)
Neoplasias Colorrectales , Tumores del Estroma Gastrointestinal , Tumores Neuroendocrinos , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/terapia , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/terapia , Humanos , Tumores Neuroendocrinos/diagnóstico , Tumores Neuroendocrinos/terapia , Tasa de Supervivencia
5.
Oncogene ; 24(20): 3285-96, 2005 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-15735702

RESUMEN

Germline mutations in the BRCA1 gene are associated with an increased susceptibility to the development of breast and ovarian cancers. Evidence suggests that BRCA1 protein plays a key role in mediating DNA damage-induced checkpoint responses. Several studies have shown that ectopic expression of BRCA1 in human cells can trigger cellular responses similar to those induced by DNA damage, including G2/M cell cycle arrest and apoptosis. While the effects of ectopic BRCA1 expression on the G2/M transition and apoptosis have been extensively studied, the factors that dictate the balance between these two responses remain poorly understood. We have recently shown that ectopic expression of BRCA1 in MCF-7 human breast cancer cells resulted in activation of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) and G2/M cell cycle arrest. Furthermore, inhibition of BRCA1-induced ERK1/2 activation using mitogen-activated protein kinase kinase 1 and 2 (MEK1/2)-specific inhibitors resulted in increased apoptosis, suggesting a potential role of ERK1/2 kinases in BRCA1-mediated G2/M checkpoint response. In this study, we assessed the role of ERK1/2 kinases in the regulation of BRCA1-mediated G2/M cell cycle arrest. Results indicate that BRCA1-induced G2/M cell cycle arrest and ERK1/2 activation correlate with changes in the level and/or activity of several key regulators of the G2/M checkpoint, including activation of Chk1 and Wee1 kinases, induction of 14-3-3, and down-regulation of Cdc25C. Furthermore, inhibition of ERK1/2 kinases using MEK1/2-specific inhibitors results in a marked attenuation of the BRCA1-induced G2/M arrest. Biochemical studies established that ERK1/2 inhibition abolished the effects of BRCA1 on components of the G2/M checkpoint, including regulation of Cdc25C expression and activation of Wee1 and Chk1 kinases. These results implicate a critical role of ERK1/2 signaling in the regulation of BRCA1 function on controlling the G2/M checkpoint responses.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas 14-3-3/metabolismo , Adenoviridae/genética , Apoptosis , Northern Blotting , Neoplasias de la Mama/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , División Celular , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , ADN/metabolismo , Daño del ADN , Regulación hacia Abajo , Activación Enzimática , Fase G2 , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Tirosina/química , Fosfatasas cdc25/metabolismo
6.
Cancer Chemother Pharmacol ; 63(5): 807-18, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18661133

RESUMEN

Previous studies have indicated that the ERK1/2 MAP kinase signaling pathway plays an important role not only in cell growth, cell cycle regulation, and differentiation, but also in determining the sensitivity of cells to anticancer agents as well. Furthermore, expression of kinase suppressor of Ras-1 (KSR1), a molecular scaffold that modulates signaling through the ERK1/2 MAP kinase pathway, has been shown to influence the cellular sensitivity to the anticancer agent cisplatin. To further define the role of KSR1 expression on drug sensitivity, the expression of KSR1 was examined in the NCI60 anticancer drug screen, a panel of cancer cell lines representing nine tissue types, established by the Developmental Therapeutics Program (DTP) at the National Cancer Institute (NCI). The expression of thousands of molecular targets has been examined in the NCI60 panel as well as the cellular toxicity for greater than 400,000 compounds. KSR1 expression varied almost 30-fold difference between the highest and lowest expressing cell lines in the NCI60. Using the COMPARE analysis algorithm, KSR1 expression was correlated with sensitivity of the compounds screened by DTP and several novel agents were identified whose sensitivity correlated with KSR1 expression in the NCI60 panel. Cytotoxicity of two agents, cytochalasin H and tunicamycin, identified through the COMPARE analysis of KSR1 expression and drug sensitivity, was also examined in wild type (KSR(+/+)) mouse embryo fibroblasts (MEFs) and MEFs deficient in KSR1 expression (KSR1(-/-)). These studies demonstrated enhanced sensitivity, as well as increased ERK activation, in KSR(-/-) MEFs following exposure to tunicamycin or cytochalasin H compared to KSR(+/+) MEFs. Furthermore, restoration of KSR1 expression in KSR(-/-) MEFs following stable transduction of cells with a KSR1 expression vector, enhanced sensitivity of cells to tunicamycin and cytochalasin H and decreased ERK1/2 activation following exposure to these drugs. In addition, the sensitivity to cytochalasin H and tunicamycin of breast cancer cell lines with low KSR1 expression, (HS578T and MDA-MB-231/ATCC), was increased relative to the sensitivity of breast cancer cells with higher levels of KSR1 (MCF7). These studies indicate that KSR1 may play an important role in the determination of cellular sensitivity to anticancer agents.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Quinasas/fisiología , Animales , Antivirales/farmacología , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Células Cultivadas , Citocalasinas/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/enzimología , Activación Enzimática/efectos de los fármacos , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Humanos , Ratones , Ratones Noqueados , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Tunicamicina/farmacología
7.
J Biol Chem ; 284(11): 6705-15, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19147494

RESUMEN

KSR1 (kinase suppressor of Ras 1) is a molecular scaffold and positive regulator of the Raf/MEK/ERK phosphorylation cascade. KSR1 is required for maximal ERK activation induced by growth factors and by some cytotoxic agents. We show here that KSR1 is also required for maximal ERK activation induced by UV light, ionizing radiation, or the DNA interstrand cross-linking agent mitomycin C (MMC). We further demonstrate a role for KSR1 in the reinitiation of the cell cycle and proliferation following cell cycle arrest induced by MMC. Cells lacking KSR1 underwent but did not recover from MMC-induced G(2)/M arrest. Expression of KSR1 allowed KSR1(-/-) cells to re-enter the cell cycle following MMC treatment. However, cells expressing a mutated form of KSR1 unable to bind ERK did not recover from MMC-induced cell cycle arrest, demonstrating the requirement for the KSR1-ERK interaction. In addition, constitutive activation of ERK was not sufficient to promote cell cycle reinitiation in MMC-treated KSR1(-/-) cells. Only cells expressing KSR1 recovered from MMC-induced cell cycle arrest. Importantly, MMC-induced DNA damage was repaired in KSR1(-/-) cells, as determined by resolution of gamma-H2AX-containing foci. These data indicate that cell cycle reinitiation is not actively signaled in the absence of KSR1, even when DNA damage has been resolved. These data reveal a specific role for the molecular scaffold KSR1 and KSR1-mediated ERK signaling in the cellular response to DNA interstrand cross-links.


Asunto(s)
Ciclo Celular/fisiología , Daño del ADN/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , Mitomicina/farmacología , Mutación , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA