Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Future Oncol ; 18(29): 3255-3266, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36000541

RESUMEN

HER2-positive gastroesophageal adenocarcinomas (GEAs) are common cancers with high mortality and the treatment options for advanced/metastatic disease are limited. Zanidatamab and tislelizumab are novel monoclonal antibodies targeting HER2 and PD-1, respectively, and have shown encouraging antitumor activity in early phase studies in multiple cancers, including GEA. Preliminary data suggest that dual targeting of the HER2 and PD-1 pathways could further improve upon the results achieved with targeting either pathway alone. Here, we describe the design of HERIZON-GEA-01, a global, randomized, open-label, active-comparator, Phase III study to evaluate and compare the efficacy and safety of zanidatamab plus chemotherapy with or without tislelizumab to the standard of care (trastuzumab plus chemotherapy) as first-line treatment for patients with advanced/metastatic HER2-positive GEAs.


HERIZON-GEA-01: A phase III study of zanidatamab + chemotherapy ± tislelizumab for first-line treatment of advanced or metastatic #HER2-positive gastroesophageal adenocarcinoma (GEA). Clinicaltrials.gov: NCT05152147. #gastriccancer #stomachcancer @ZymeworksInc @BeiGeneGlobal Clinical Trial Registration: NCT05152147 (ClinicalTrials.gov).


Asunto(s)
Adenocarcinoma , Antineoplásicos , Neoplasias Esofágicas , Neoplasias Gástricas , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Unión Esofagogástrica/patología , Humanos , Receptor de Muerte Celular Programada 1 , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/patología , Trastuzumab/farmacología , Trastuzumab/uso terapéutico
2.
Blood Res ; 55(1): 57-61, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32269976

RESUMEN

BACKGROUND: Autologous stem cell transplantation (autoSCT) can extend remission of mantle cell lymphoma (MCL), but the management of subsequent relapse is challenging. METHODS: We examined consecutive patients with MCL who underwent autoSCT at Veterans Affairs Puget Sound Health Care System between 2009 and 2017 (N=37). RESULTS: Ten patients experienced disease progression after autoSCT and were included in this analysis. Median progression free survival after autoSCT was 1.8 years (range, 0.3-7.1) and median overall survival after progression was only 0.7 years (range, 0.1 to not reached). The 3 patients who survived more than 1 year after progression were treated with ibrutinib. CONCLUSION: Our findings suggest that ibrutinib can achieve relatively prolonged control of MCL progressing after autoSCT.

3.
Sci Rep ; 9(1): 5488, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940846

RESUMEN

Insertional mutagenesis is a powerful means of identifying cancer drivers in animal models. We used the Sleeping Beauty (SB) transposon/transposase system to identify activated oncogenes in hematologic cancers in wild-type mice and mice that express a stabilized cyclin E protein (termed cyclin ET74AT393A). Cyclin E governs cell division and is misregulated in human cancers. Cyclin ET74AT393A mice develop ineffective erythropoiesis that resembles early-stage human myelodysplastic syndrome, and we sought to identify oncogenes that might cooperate with cyclin E hyperactivity in leukemogenesis. SB activation in hematopoietic precursors caused T-cell leukemia/lymphomas (T-ALL) and pure red blood cell erythroleukemias (EL). Analysis of >12,000 SB integration sites revealed markedly different oncogene activations in EL and T-ALL: Notch1 and Ikaros were most common in T-ALL, whereas ETS transcription factors (Erg and Ets1) were targeted in most ELs. Cyclin E status did not impact leukemogenesis or oncogene activations. Whereas most SB insertions were lost during culture of EL cell lines, Erg insertions were retained, indicating Erg's key role in these neoplasms. Surprisingly, cyclin ET74AT393A conferred growth factor independence and altered Erg-dependent differentiation in EL cell lines. These studies provide new molecular insights into erythroid leukemia and suggest potential therapeutic targets for human leukemia.


Asunto(s)
Ciclina E/genética , Leucemia Eritroblástica Aguda/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Transposasas/genética , Animales , Técnicas de Cultivo de Célula , Elementos Transponibles de ADN , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Ratones , Mutagénesis Insercional , Proteínas Oncogénicas/genética , Regulador Transcripcional ERG/genética
4.
Proc Natl Acad Sci U S A ; 115(21): 5462-5467, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29735700

RESUMEN

The Fbw7 (F-box/WD repeat-containing protein 7) ubiquitin ligase targets multiple oncoproteins for degradation and is commonly mutated in cancers. Like other pleiotropic tumor suppressors, Fbw7's complex biology has impeded our understanding of how Fbw7 mutations promote tumorigenesis and hindered the development of targeted therapies. To address these needs, we employed a transfer learning approach to derive gene-expression signatures from The Cancer Gene Atlas datasets that predict Fbw7 mutational status across tumor types and identified the pathways enriched within these signatures. Genes involved in mitochondrial function were highly enriched in pan-cancer signatures that predict Fbw7 mutations. Studies in isogenic colorectal cancer cell lines that differed in Fbw7 mutational status confirmed that Fbw7 mutations increase mitochondrial gene expression. Surprisingly, Fbw7 mutations shifted cellular metabolism toward oxidative phosphorylation and caused context-specific metabolic vulnerabilities. Our approach revealed unexpected metabolic reprogramming and possible therapeutic targets in Fbw7-mutant cancers and provides a framework to study other complex, oncogenic mutations.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Metaboloma , Mitocondrias/metabolismo , Mutación , Respiración de la Célula , Neoplasias Colorrectales/genética , Perfilación de la Expresión Génica , Humanos , Mitocondrias/patología , Fosforilación Oxidativa , Estrés Oxidativo , Fosforilación , Ubiquitina , Ubiquitinación
5.
Transpl Infect Dis ; 19(3)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28295973

RESUMEN

Alternaria species have been reported as a rare cause of fungal infection in organ and stem cell transplant recipients, but to date, no reports have been published of infection in humans caused by Alternaria rosae. Here, we report cutaneous A. rosae infection in a 66-year-old farmer with a history of primary myelofibrosis who had undergone allogeneic unrelated donor hematopoietic stem cell transplantation. Forty-nine days post transplant, he presented with a nodule on the thumb with no findings suggestive of disseminated infection. Pathology, culture, and molecular speciation showed the nodule was caused by cutaneous A. rosae. He had been on voriconazole as antifungal prophylaxis, but was found to have a subtherapeutic voriconazole level. He was switched to posaconazole based on published in vitro data showing its superior efficacy in Alternaria treatment. Susceptibility testing showed that the A. rosae isolate was indeed susceptible to posaconazole. His cutaneous lesion remained stable, but he died from respiratory failure secondary to lobar pneumonia. At lung autopsy, A. rosae was not identified in the lungs. We believe this to be the first published report, to our knowledge, of A. rosae infection in humans.


Asunto(s)
Alternaria/patogenicidad , Alternariosis/microbiología , Antifúngicos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Feohifomicosis/microbiología , Mielofibrosis Primaria/terapia , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/terapia , Aciclovir/uso terapéutico , Anciano , Alternaria/aislamiento & purificación , Profilaxis Antibiótica/métodos , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/métodos , Resultado Fatal , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Mano/diagnóstico por imagen , Humanos , Terapia de Inmunosupresión/efectos adversos , Terapia de Inmunosupresión/métodos , Levofloxacino/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Pruebas de Sensibilidad Microbiana , Senos Paranasales/diagnóstico por imagen , Neumonía/complicaciones , Prednisona/uso terapéutico , Insuficiencia Respiratoria/complicaciones , Esporas Fúngicas/aislamiento & purificación , Esporas Fúngicas/patogenicidad , Trasplante Homólogo/efectos adversos , Triazoles/uso terapéutico , Voriconazol/uso terapéutico
6.
Gastroenterol Rep (Oxf) ; 3(4): 316-29, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26510453

RESUMEN

Gastroesophageal cancer is a significant global problem that frequently presents at an incurable stage and has very poor survival with standard chemotherapy approaches. This review will examine the epidemiology and molecular biology of gastroesophageal cancer and will focus on the key deregulated signaling pathways that have been targeted in the clinic. A comprehensive overview of clinical data highlighting successes and failures with targeted agents will be presented. Most notably, HER2-targeted therapy with the monoclonal antibody trastuzumab has proven beneficial in first-line therapy and has been incorporated into standard practice. Targeting the VEGF pathway has also proven beneficial, and the VEGFR-targeted monoclonal antibody ramucirumab is now approved for second-line therapy. In contrast to these positive results, agents targeting the EGFR and MET pathways have been evaluated extensively in gastroesophageal cancer but have repeatedly failed to show benefit. An increased understanding of the molecular predictors of response to targeted therapies is sorely needed. In the future, improved molecular pathology approaches should subdivide this heterogeneous disease entity to allow individualization of cancer therapy based on integrated and global identification of deregulated signaling pathways. Better patient selection, rational combinations of targeted therapies and incorporation of emerging immunotherapeutic approaches should further improve the treatment of this deadly disease.

9.
Genes Dev ; 27(23): 2531-6, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24298052

RESUMEN

The Fbw7 tumor suppressor targets a broad network of proteins for ubiquitylation. Here we show critical functions for Fbw7 dimerization in regulating the specificity and robustness of degradation. Dimerization enables Fbw7 to target substrates through concerted binding to two suboptimal and independent recognition sites. Accordingly, an endogenous dimerization-deficient Fbw7 mutation stabilizes suboptimal substrates. Dimerization increases Fbw7's robustness by preserving its function in the setting of mutations that disable Fbw7 monomers, thereby buffering against pathogenic mutations. Finally, dimerization regulates Fbw7 stability, and this likely involves Fbw7 trans-autoubiquitylation. Our study reveals novel functions of Fbw7 dimerization and an unanticipated complexity in substrate degradation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/química , Dimerización , Proteínas F-Box/química , Proteína 7 que Contiene Repeticiones F-Box-WD , Células HCT116 , Humanos , Ligasas/química , Ligasas/metabolismo , Mutación , Unión Proteica , Estabilidad Proteica , Especificidad por Sustrato , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
10.
Mol Cell Biol ; 32(11): 2160-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473991

RESUMEN

Colorectal cancer (CRC) remains a major cause of cancer mortality worldwide. Murine models have yielded critical insights into CRC pathogenesis, but they often fail to recapitulate advanced-disease phenotypes, notably metastasis and chromosomal instability (CIN). New models are thus needed to understand disease progression and to develop therapies. We sought to model advanced CRC by inactivating two tumor suppressors that are mutated in human CRCs, the Fbw7 ubiquitin ligase and p53. Here we report that Fbw7 deletion alters differentiation and proliferation in the gut epithelium and stabilizes oncogenic Fbw7 substrates, such as cyclin E and Myc. However, Fbw7 deletion does not cause tumorigenesis in the gut. In contrast, codeletion of both Fbw7 and p53 causes highly penetrant, aggressive, and metastatic adenocarcinomas, and allografts derived from these tumors form highly malignant adenocarcinomas. In vitro evidence indicates that Fbw7 ablation promotes genetic instability that is suppressed by p53, and we show that most Fbw7⁻/⁻; p53⁻/⁻ carcinomas exhibit a CIN⁺ phenotype. We conclude that Fbw7 and p53 synergistically suppress adenocarcinomas that mimic advanced human CRC with respect to histopathology, metastasis, and CIN. This model thus represents a novel tool for studies of advanced CRC as well as carcinogenesis associated with ubiquitin pathway mutations.


Asunto(s)
Adenocarcinoma , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales , Modelos Animales de Enfermedad , Proteínas F-Box/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Transformación Celular Neoplásica , Inestabilidad Cromosómica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
11.
Blood ; 119(11): 2657-64, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22234678

RESUMEN

Studies by the International Working Group showed that the prognosis of myelofibrosis patients is predicted by the Dynamic International Prognostic Scoring System (DIPSS) risk categorization, which includes patient age, constitutional symptoms, hemoglobin, leukocyte count, and circulating blasts. We evaluated the prognostic usefulness of the DIPSS in 170 patients with myelofibrosis, 12 to 78 years of age (median, 51.5 years of age), who received hematopoietic cell transplantation (HCT) between 1990 and 2009 from related (n = 86) or unrelated donors (n = 84). By DIPSS, 21 patients had low-risk disease, 48 had intermediate-1, 50 had intermediate-2, and 51 had high-risk disease. Five-year incidence of relapse, relapse-free survival, overall survival, and nonrelapse mortality for all patients were 10%, 57%, 57%, and 34%, respectively. Among patients with DIPSS high-risk disease, the hazard ratio for post-HCT mortality was 4.11 (95% CI, 1.44-11.78; P = .008), and for nonrelapse mortality was 3.41 (95% CI, 1.15-10.09; P = .03) compared with low-risk patients. After a median follow-up of 5.9 years, the median survivals have not been reached for DIPSS risk groups low and intermediate-1, and were 7 and 2.5 years for intermediate-2 and high-risk patients, respectively. Thus, HCT was curative for a large proportion of patients with myelofibrosis, and post-HCT success was dependent on pre-HCT DIPSS classification.


Asunto(s)
Modelos Estadísticos , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/terapia , Trasplante de Células Madre de Sangre Periférica , Mielofibrosis Primaria/mortalidad , Mielofibrosis Primaria/terapia , Adolescente , Adulto , Anciano , Niño , Femenino , Hemoglobinas/análisis , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Mielofibrosis Primaria/sangre , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia , Adulto Joven
12.
J Cell Biol ; 181(6): 913-20, 2008 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-18559665

RESUMEN

The SCF(FBW7) ubiquitin ligase degrades proteins involved in cell division, growth, and differentiation and is commonly mutated in cancers. The Fbw7 locus encodes three protein isoforms that occupy distinct subcellular localizations, suggesting that each has unique functions. We used gene targeting to create isoform-specific Fbw7-null mutations in human cells and found that the nucleoplasmic Fbw7alpha isoform accounts for almost all Fbw7 activity toward cyclin E, c-Myc, and sterol regulatory element binding protein 1. Cyclin E sensitivity to Fbw7 varies during the cell cycle, and this correlates with changes in cyclin E-cyclin-dependent kinase 2 (CDK2)-specific activity, cyclin E autophosphorylation, and CDK2 inhibitory phosphorylation. These data suggest that oscillations in cyclin E-CDK2-specific activity during the cell cycle regulate the timing of cyclin E degradation. Moreover, they highlight the utility of adeno-associated virus-mediated gene targeting in functional analyses of complex loci.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proteínas F-Box/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Estabilidad de Enzimas , Proteína 7 que Contiene Repeticiones F-Box-WD , Marcación de Gen , Humanos , Isoenzimas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Especificidad por Sustrato
13.
J Exp Med ; 204(8): 1813-24, 2007 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-17646409

RESUMEN

gamma-secretase inhibitors (GSIs) can block NOTCH receptor signaling in vitro and therefore offer an attractive targeted therapy for tumors dependent on deregulated NOTCH activity. To clarify the basis for GSI resistance in T cell acute lymphoblastic leukemia (T-ALL), we studied T-ALL cell lines with constitutive expression of the NOTCH intracellular domain (NICD), but that lacked C-terminal truncating mutations in NOTCH1. Each of the seven cell lines examined and 7 of 81 (8.6%) primary T-ALL samples harbored either a mutation or homozygous deletion of the gene FBW7, a ubiquitin ligase implicated in NICD turnover. Indeed, we show that FBW7 mutants cannot bind to the NICD and define the phosphodegron region of the NICD required for FBW7 binding. Although the mutant forms of FBW7 were still able to bind to MYC, they do not target it for degradation, suggesting that stabilization of both NICD and its principle downstream target, MYC, may contribute to transformation in leukemias with FBW7 mutations. In addition, we show that all seven leukemic cell lines with FBW7 mutations were resistant to the MRK-003 GSI. Most of these resistant lines also failed to down-regulate the mRNA levels of the NOTCH targets MYC and DELTEX1 after treatment with MRK-003, implying that residual NOTCH signaling in T-ALLs with FBW7 mutations contributes to GSI resistance.


Asunto(s)
Proteínas de Ciclo Celular/genética , Inhibidores Enzimáticos/farmacología , Proteínas F-Box/genética , Regulación Neoplásica de la Expresión Génica , Leucemia/genética , Leucemia/metabolismo , Mutación , Receptores Notch/genética , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptores Notch/metabolismo
14.
Cell Tissue Res ; 325(2): 263-76, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16557385

RESUMEN

Mechanisms controlling epithelial proliferation and differentiation in the prostate have been primarily investigated in mouse models. The regulation of proliferation and differentiation is poorly understood in human prostate epithelial cells. In vivo, the glandular prostate epithelium consists of a p63-positive proliferating basal cell layer and a post-mitotic p27-positive secretory cell layer. We have established an organized stratified culture system of human primary prostate epithelial cells to gain insight into mechanisms regulating proliferation and differentiation. In this system, expression of p63 is observed in the bottom layer. In addition, BrdU incorporation persists even though cells are confluent. In contrast, in the upper layer, p63 expression is greatly diminished, p27 is expressed, and the cells are growth arrested. Overexpression of cyclin D1 or knockdown of p27 does not increase proliferation. After inactivation of the nuclear phosphoprotein Rb, the cell layers remain organized and cell proliferation increases only in the bottom layer. Furthermore, the expression of p63 remains confined to the bottom layer after Rb inactivation. Altogether, this in vitro model recapitulates certain aspects of in vivo growth regulation and differentiation and suggests that the loss of Rb family proteins in human cells trigger hyperplasia but is not sufficient for transformation.


Asunto(s)
Proliferación Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Próstata/citología , Técnicas de Cultivo de Tejidos , Diferenciación Celular/fisiología , Ciclina D1/genética , Ciclina D1/metabolismo , Técnicas de Transferencia de Gen , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Proteínas E7 de Papillomavirus/biosíntesis , Proteínas E7 de Papillomavirus/genética , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética
15.
Prostate ; 65(2): 141-50, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15924336

RESUMEN

BACKGROUND: The present study was designed to determine if estrogens change microtubule polymerization and modulate cell cycle progression in vitro, related to modulation of tubulin expression and to determine if estrogens had antagonistic or synergistic effects with microtubule active agents. METHODS: cDNA array analysis of LNCaP cells treated with the estrogens, estradiol, estrone, diethylstilbestrol (DES), and 2-methoxyestradiol (2-ME) was carried out and the results confirmed by PCR and Western blotting. Microtubule arrays in cells treated with estrogens were assessed using indirect immunofluorescence. The effects of combining estrogens with taxane was assessed by MTT assay and flow cytometry for cell cycle kinetics. Human prostate cancer xenografts were treated with DES and docetaxel to assess the effects of combining estrogens and taxane in vivo. RESULTS: Treatment of LNCaP cells with DES and 2-ME suppressed transcripts and protein for beta-tubulin isotype IVa. This effect on tubulin synthesis was not blocked by estrogen or androgen receptor modulators. Other estrogens had no effect on beta-tubulin expression. 2-ME and DES decreased the density of microtubules. The administration of DES or 2-ME with paclitaxel enhanced cytotoxicity and G(2)-M arrest in vitro. DES enhanced tumor suppression in a human prostate cancer xenograft model when combined with the taxane docetaxel. CONCLUSION: The use of DES and 2-ME enhances the effects of taxanes and may be a novel and important means of increasing therapeutic efficacy of cytotoxic chemotherapy against prostate carcinoma.


Asunto(s)
Estrógenos/farmacología , Neoplasias de la Próstata/patología , Taxoides/efectos adversos , Tubulina (Proteína)/biosíntesis , Western Blotting , Ciclo Celular/fisiología , Citometría de Flujo , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
16.
Curr Biol ; 14(20): 1852-7, 2004 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-15498494

RESUMEN

The human tumor suppressor Fbw7/hCdc4 functions as a phosphoepitope-specific substrate recognition component of SCF ubiquitin ligases that catalyzes the ubiquitination of cyclin E , Notch , c-Jun and c-Myc . Fbw7 loss in cancer may thus have profound effects on the pathways that govern cell division, differentiation, apoptosis, and cell growth. Fbw7-inactivating mutations occur in human tumor cell lines and primary cancers , and Fbw7 loss in cultured cells causes genetic instability . In mice, deletion of Fbw7 leads to embryonic lethality associated with defective Notch and cyclin E regulation . The human Fbw7 locus encodes three protein isoforms (Fbw7alpha, Fbw7beta, and Fbw7gamma) . We find that these isoforms occupy discrete subcellular compartments and have identified cis-acting localization signals within each isoform. Surprisingly, the Fbw7gamma isoform is nucleolar, colocalizes with c-Myc when the proteasome is inhibited, and regulates nucleolar c-Myc accumulation. Moreover, we find that knockdown of Fbw7 increases cell size consistent with its ability to control c-Myc levels in the nucleolus. We suggest that interactions between c-Myc and Fbw7gamma within the nucleolus regulate c-Myc's growth-promoting function and that c-Myc activation is likely to be an important oncogenic consequence of Fbw7 loss in cancers.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Ciclo Celular/genética , Nucléolo Celular/metabolismo , Tamaño de la Célula , Células Cultivadas , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Técnicas de Transferencia de Gen , Humanos , Immunoblotting , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina-Proteína Ligasas/genética
17.
Mol Cell Biol ; 24(20): 8813-22, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456857

RESUMEN

Although previous studies demonstrate that appropriate Notch signaling is required during angiogenesis and in vascular homeostasis, the mechanisms by which Notch regulates vascular function remain to be elucidated. Here, we show that activation of the Notch pathway by the ligand Jagged1 reduces the proliferation of endothelial cells. Notch activation inhibits proliferation of endothelial cells in a cell-autonomous manner by inhibiting phosphorylation of the retinoblastoma protein (Rb). During cell cycle entry, p21Cip1 is upregulated in endothelial cells. Activated Notch inhibits mitogen-induced upregulation of p21Cip1 and delays cyclin D-cdk4-mediated Rb phosphorylation. Notch-dependent repression of p21Cip1 prevents nuclear localization of cyclin D and cdk4. The necessity of p21Cip1 for nuclear translocation of cyclin D-cdk4 and S-phase entry in endothelial cells was demonstrated by targeted downregulation of p21Cip1 by using RNA interference. We further demonstrate that when endothelial cells reach confluence, Notch is activated and p21Cip1 is downregulated. Inhibition of the Notch pathway at confluence prevents p21Cip1 downregulation and induces Rb phosphorylation. We suggest that Notch activation contributes to contact inhibition of endothelial cells, in part through repression of p21Cip1 expression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Inhibición de Contacto/fisiología , Células Endoteliales/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas de Unión al Calcio , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Ciclina D , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Células Endoteliales/citología , Endotelio Vascular/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosforilación , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Receptor Notch1 , Receptor Notch4 , Receptores de Superficie Celular/genética , Receptores Notch , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas Serrate-Jagged , Factores de Transcripción/genética
18.
Proc Natl Acad Sci U S A ; 101(24): 9085-90, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15150404

RESUMEN

Myc proteins regulate cell growth and division and are implicated in a wide range of human cancers. We show here that Fbw7, a component of the SCF(Fbw7) ubiquitin ligase and a tumor suppressor, promotes proteasome-dependent c-Myc turnover in vivo and c-Myc ubiquitination in vitro. Phosphorylation of c-Myc on threonine-58 (T58) by glycogen synthase kinase 3 regulates the binding of Fbw7 to c-Myc as well as Fbw7-mediated c-Myc degradation and ubiquitination. T58 is the most frequent site of c-myc mutations in lymphoma cells, and our findings suggest that c-Myc activation is one of the key oncogenic consequences of Fbw7 loss in cancer. Because Fbw7 mediates the degradation of cyclin E, Notch, and c-Jun, as well as c-Myc, the loss of Fbw7 is likely to elicit profound effects on cell proliferation during tumorigenesis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/genética , Línea Celular , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Fibroblastos/metabolismo , Células HeLa , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transducción Genética , Transfección , Proteínas Supresoras de Tumor/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
19.
Mol Cell ; 12(2): 381-92, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14536078

RESUMEN

Autophosphorylation-triggered ubiquitination has been proposed to be the major pathway regulating cyclin E protein abundance: phosphorylation of cyclin E on T380 by its associated CDK allows binding to the receptor subunit, Fbw7, of the SCFFbw7 ubiquitin ligase. We have tested this model in vivo and found it to be an inadequate representation of the pathways that regulate cyclin E degradation. We show that assembly of cyclin E into cyclin E-Cdk2 complexes is required in vivo for turnover by the Fbw7 pathway; that Cdk2 activity is required for cyclin E turnover in vivo because it phosphorylates S384; that phosphorylation of T380 in vivo does not require Cdk2 and is mediated primarily by GSK3; and that two additional phosphorylation sites, T62 and S372, are also required for turnover. Thus, cyclin E turnover is controlled by multiple biological inputs and cannot be understood in terms of autophosphorylation alone.


Asunto(s)
Quinasas CDC2-CDC28/fisiología , Ciclina E/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Animales , Sitios de Unión , Quinasas CDC2-CDC28/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasas/metabolismo , Humanos , Immunoblotting , Ratones , Mutación , Fosforilación , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Retroviridae/metabolismo , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/metabolismo
20.
Trends Cell Biol ; 13(8): 396-9, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12888290

RESUMEN

Cyclin-dependent kinase 2 (CDK2) regulates diverse aspects of the mammalian cell cycle. Most cancer cells contain mutations in the pathways that control CDK2, and CDK2 activity has received much attention as a target for cancer therapy. However, a recent report demonstrating that some cancer cells can proliferate without CDK2 activity questions the essential role of CDK2 in cell-cycle control, as well as its suitability as a therapeutic target.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Ciclo Celular/fisiología , Proteínas de Unión al ADN , Proteínas Proto-Oncogénicas , Proteínas de Saccharomyces cerevisiae , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Factores de Transcripción E2F , Fase G1/fisiología , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosforilación , Proteína de Retinoblastoma/metabolismo , Factor de Transcripción DP1 , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA