Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Osteoarthritis Cartilage ; 29(3): 402-412, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33227437

RESUMEN

OBJECTIVE: Cellular senescence is a phenotypic state characterized by stable cell-cycle arrest, enhanced lysosomal activity, and the secretion of inflammatory molecules and matrix degrading enzymes. Senescence has been implicated in osteoarthritis (OA) pathophysiology; however, the mechanisms that drive senescence induction in cartilage and other joint tissues are unknown. While numerous physiological signals are capable of initiating senescence, one emerging theme is that damaged cells convert to senescence in response to sustained mitogenic stimulation. The goal of this study was to develop an in vitro articular cartilage explant model to investigate the mechanisms of senescence induction. DESIGN: This study utilized healthy cartilage derived from cadaveric equine stifles and human ankles. Explants were irradiated to initiate DNA damage, and mitogenic stimulation was provided through serum-containing medium and treatment with transforming growth factor ß1 and basic fibroblastic growth factor. Readouts of senescence were a quantitative flow cytometry assay to detect senescence-associated ß galactosidase activity (SA-ß-gal), immunofluorescence for p16 and γH2AX, and qPCR for the expression of inflammatory genes. RESULTS: Human cartilage explants required both irradiation and mitogenic stimulation to induce senescence as compared to baseline control conditions (7.16% vs 2.34% SA-ß-gal high, p = 0.0007). These conditions also resulted in chondrocyte clusters within explants, a persistent DNA damage response, increased p16, and gene expression changes. CONCLUSIONS: Treatment of cartilage explants with mitogenic stimuli in the context of cellular damage reliably induces high levels of SA-ß-gal activity and other senescence markers, which provides a physiologically relevant model system to investigate the mechanisms of senescence induction.


Asunto(s)
Cartílago Articular/metabolismo , Senescencia Celular/genética , Condrocitos/metabolismo , Animales , Articulación del Tobillo , Cartílago Articular/citología , Cartílago Articular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Condrocitos/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica/efectos de los fármacos , Histonas/efectos de los fármacos , Histonas/metabolismo , Caballos , Humanos , Técnicas In Vitro , Inflamación/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/efectos de los fármacos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Interleucina-6/genética , Metaloproteinasa 13 de la Matriz/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/genética , Mitógenos/farmacología , Rodilla de Cuadrúpedos , Factor de Crecimiento Transformador beta1/farmacología , beta-Galactosidasa/efectos de los fármacos , beta-Galactosidasa/metabolismo
2.
Arch Gynecol Obstet ; 303(2): 501-511, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33219482

RESUMEN

PURPOSE: The effect of SAM vaginal gel, a medical device containing adsorptive silicon dioxide and antioxidative sodium selenite and citric acid, on histologically-proven cervical intraepithelial neoplasia type 2 (CIN2) as well as p16 positive CIN1, and on the presence of the onco-marker p16 was investigated. METHODS: 216 women aged 25-60 years were randomized to either receive an intravaginal daily dose of SAM gel for three 28-day periods, or be followed-up without intervention. The primary endpoint was efficacy, defined as a combined histological and cytological regression. At baseline and after 3 months participants had: a guided biopsy including p16 immunohistochemical (IHC) staining, only if a lesion was visible at colposcopy; a cervical smear for cytology, high-risk human papillomavirus (hr-HPV) and a p16/Ki-67 test. At 6 months a further cytology and p16/Ki-67 test was performed. RESULTS: Regression of CIN lesions was observed in 78 out of 108 patients (72.2%) in the SAM gel arm and in 27 out of 108 patients (25.0%) in the control arm. Similarly, the change in the p16/Ki-67 cytological test status was significantly in favor of the treatment arm. The prevalence of hr-HPV decreased significantly (p < 0.001) in the treatment arm, from 87.0% to 39.8%, while it slightly increased in the control arm, from 78.7% to 83.3%. At 6 months the cytological regression in the treatment group and the highly significant effect on p16/Ki-67 was still present. CONCLUSION: SAM vaginal gel enhances the regression of cervical lesions and clears hr-HPV and p16/Ki-67 in smears significantly, thus offering an active non-destructive management to prevent cervical cancer. TRIAL REGISTRATION NUMBER: ISRCTN11009040, date of registration: 10/12/2019; https://doi.org/10.1186/ISRCTN11009040 ; retrospectively registered.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Antígeno Ki-67/efectos de los fármacos , Infecciones por Papillomavirus/terapia , Dióxido de Silicio/administración & dosificación , Selenito de Sodio/administración & dosificación , Displasia del Cuello del Útero/terapia , Cremas, Espumas y Geles Vaginales , Administración Intravaginal , Adulto , Antioxidantes/administración & dosificación , Biomarcadores de Tumor/análisis , Ácido Cítrico/administración & dosificación , Colposcopía , Citodiagnóstico , Femenino , Genes p16 , Humanos , Persona de Mediana Edad , Infecciones por Papillomavirus/virología , Embarazo , Resultado del Tratamiento , Neoplasias del Cuello Uterino/prevención & control , Frotis Vaginal , Displasia del Cuello del Útero/patología
3.
Clin Epigenetics ; 12(1): 142, 2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32958049

RESUMEN

BACKGROUND: Epigenetic reprogramming using DNA demethylating drugs is a promising approach for cancer therapy, but its efficacy is highly dependent on the dosing regimen. Low-dose treatment for a prolonged period shows a remarkable therapeutic efficacy, despite its small demethylating effect. Here, we aimed to explore the mechanisms of how such low-dose treatment shows this remarkable efficacy by focusing on epigenetic reprograming at the single-cell level. METHODS: Expression profiles in HCT116 cells treated with decitabine (DAC) were analyzed by single-cell RNA-sequencing (scRNA-seq). Functional consequences and DNA demethylation at the single-cell level were analyzed using cloned HCT116 cells after DAC treatment. RESULTS: scRNA-seq revealed that DAC-treated cells had highly diverse expression profiles at the single-cell level, and tumor-suppressor genes, endogenous retroviruses, and interferon-stimulated genes were upregulated in random fractions of cells. DNA methylation analysis of cloned HCT116 cells revealed that, while only partial reduction of DNA methylation levels was observed in bulk cells, complete demethylation of specific cancer-related genes, such as cell cycle regulation, WNT pathway, p53 pathway, and TGF-ß pathway, was observed, depending upon clones. Functionally, a clone with complete demethylation of CDKN2A (p16) had a larger fraction of cells with tetraploid than parental cells, indicating induction of cellular senescence due to normalization of cell cycle regulation. CONCLUSIONS: Epigenetic reprogramming of specific cancer-related pathways at the single-cell level is likely to underlie the remarkable efficacy of low-dose DNA demethylating therapy.


Asunto(s)
Reprogramación Celular/genética , Metilación de ADN/efectos de los fármacos , Epigenómica/métodos , Células HCT116/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Análisis de la Célula Individual/métodos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Desmetilación del ADN , Decitabina/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes p53/efectos de los fármacos , Células HCT116/metabolismo , Humanos , Neoplasias/genética , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
4.
Aging (Albany NY) ; 12(24): 26047-26062, 2020 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-33401248

RESUMEN

6-Bromoindirubin-3'-oxime (6BIO) is a novel small molecule that exerts positive effects on several age-related alterations. However, the anti-aging effects of 6BIO on the aging heart remain unknown. Herein, we aim to investigate the effects of 6BIO on the myocardium and its underlying mechanism in vivo and vitro. Following 6BIO treatment, an increased p53 contents, a reduced p16 and ß-gal levels, and attenuation of cardiac fibrosis were observed, suggesting 6BIO retarded aging of cardiomyocytes. As observed, 6BIO reduced p62 contents, elevated the levels of Beclin-1 and the ratio of LC3II/I, indicating the induction of autophagy, while the reduction of the accumulation of ROS indicated 6BIO alleviated oxidative stress. In addition, 6BIO treatment inhibited both GSK3ß signaling and mTOR signaling. 6BIO might be a promising agent for preventing myocardium from aging.


Asunto(s)
Envejecimiento/efectos de los fármacos , Autofagia/efectos de los fármacos , Corazón/efectos de los fármacos , Indoles/farmacología , Mioblastos Cardíacos/efectos de los fármacos , Miocardio/metabolismo , Oximas/farmacología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Antioxidantes/metabolismo , Beclina-1/efectos de los fármacos , Beclina-1/metabolismo , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibrosis , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ratones , Mioblastos Cardíacos/metabolismo , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/farmacología , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , beta-Galactosidasa/efectos de los fármacos , beta-Galactosidasa/metabolismo
5.
J Gynecol Oncol ; 30(2): e26, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30740957

RESUMEN

OBJECTIVE: Poly (ADP-ribose) polymerase (PARP) is an important molecule in the early stress response of DNA damage, which is involved in DNA damage repair and cellular senescence. Olaparib, as PARP inhibitor, has an anti-tumor effect on high grade serous ovarian cancer, but its effects on cellular senescence have not been reported. This study intends to explore the role of olaparib in the regulation of senescence in ovarian cancer cells. METHODS: The effects of olaparib on the senescence of ovarian cancer cells were detected by using the senescence-associated ß-galactosidase (SA-ß-Gal) and senescence-associated heterochromatin aggregation (SAHF). Quantitative real-time polymerase chain reaction was used to analyze the senescence-associated secretory phenotype (SASP). Cell cycle and apoptosis were detected by flow cytometry. The effect of olaparib on tumor growth was analyzed in a nude mouse xenograft transplantation model. RESULTS: Long-term (6 days) treatment with olaparib (5 µ) significantly inhibited the growth of ovarian cancer cells, leading to arrest the cell cycle at G0/G1 phase, significant increase the number of positive SA-ß-Gal stained cells and positive SAHF cells. The expression of P16 and retinoblastoma protein (p-RB) were significantly enhanced in SKOV3 cells under olaparib treated, meanwhile, the expression of P53 and p-RB were upregulated in A2780 cells. In OVCAR-3 cells, the expression of P53 was downregulated and p-RB was upregulated. Mice with SKOV3 xenograft transplantation was given olaparib (10 mg/kg/day) via abdominal cavity administration, the tumor volume was reduced (p<0.01). CONCLUSION: Continuous low dosage administration of olaparib induced senescence under P16 or P53 dependent manner in ovarian cancer.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Neoplasias Ováricas/patología , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Regulación hacia Abajo , Femenino , Humanos , Ratones Desnudos , Neoplasias Experimentales , Proteína de Retinoblastoma/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
6.
Nat Med ; 23(4): 483-492, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28263309

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumor that is located in the pons and primarily affects children. Nearly 80% of DIPGs harbor mutations in histone H3 genes, wherein lysine 27 is substituted with methionine (H3K27M). H3K27M has been shown to inhibit polycomb repressive complex 2 (PRC2), a multiprotein complex responsible for the methylation of H3 at lysine 27 (H3K27me), by binding to its catalytic subunit EZH2. Although DIPGs with the H3K27M mutation show global loss of H3K27me3, several genes retain H3K27me3. Here we describe a mouse model of DIPG in which H3K27M potentiates tumorigenesis. Using this model and primary patient-derived DIPG cell lines, we show that H3K27M-expressing tumors require PRC2 for proliferation. Furthermore, we demonstrate that small-molecule EZH2 inhibitors abolish tumor cell growth through a mechanism that is dependent on the induction of the tumor-suppressor protein p16INK4A. Genome-wide enrichment analyses show that the genes that retain H3K27me3 in H3K27M cells are strong polycomb targets. Furthermore, we find a highly significant overlap between genes that retain H3K27me3 in the DIPG mouse model and in human primary DIPGs expressing H3K27M. Taken together, these results show that residual PRC2 activity is required for the proliferation of H3K27M-expressing DIPGs, and that inhibition of EZH2 is a potential therapeutic strategy for the treatment of these tumors.


Asunto(s)
Neoplasias del Tronco Encefálico/genética , Proliferación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Glioma/genética , Histonas/genética , Animales , Benzamidas/farmacología , Compuestos de Bifenilo , Neoplasias Encefálicas/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cromatografía Liquida , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Técnicas de Inactivación de Genes , Glioblastoma/genética , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Indazoles/farmacología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Morfolinas , Mutación , Trasplante de Neoplasias , Células-Madre Neurales , Complejo Represivo Polycomb 2/genética , Piridonas/farmacología , Espectrometría de Masas en Tándem , Proteína p14ARF Supresora de Tumor/efectos de los fármacos , Proteína p14ARF Supresora de Tumor/genética
7.
Nat Med ; 22(1): 78-83, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26657143

RESUMEN

Senescent cells (SCs) accumulate with age and after genotoxic stress, such as total-body irradiation (TBI). Clearance of SCs in a progeroid mouse model using a transgenic approach delays several age-associated disorders, suggesting that SCs play a causative role in certain age-related pathologies. Thus, a 'senolytic' pharmacological agent that can selectively kill SCs holds promise for rejuvenating tissue stem cells and extending health span. To test this idea, we screened a collection of compounds and identified ABT263 (a specific inhibitor of the anti-apoptotic proteins BCL-2 and BCL-xL) as a potent senolytic drug. We show that ABT263 selectively kills SCs in culture in a cell type- and species-independent manner by inducing apoptosis. Oral administration of ABT263 to either sublethally irradiated or normally aged mice effectively depleted SCs, including senescent bone marrow hematopoietic stem cells (HSCs) and senescent muscle stem cells (MuSCs). Notably, this depletion mitigated TBI-induced premature aging of the hematopoietic system and rejuvenated the aged HSCs and MuSCs in normally aged mice. Our results demonstrate that selective clearance of SCs by a pharmacological agent is beneficial in part through its rejuvenation of aged tissue stem cells. Thus, senolytic drugs may represent a new class of radiation mitigators and anti-aging agents.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Senescencia Celular , Células Madre Hematopoyéticas/efectos de los fármacos , Mioblastos/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Antivirales/farmacología , Linfocitos B/efectos de los fármacos , Western Blotting , Ciclo Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Ganciclovir/farmacología , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Microscopía , Músculo Esquelético/citología , Mioblastos/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Rejuvenecimiento , Irradiación Corporal Total , Proteína bcl-X/genética
8.
Toxicol In Vitro ; 29(1): 242-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25448810

RESUMEN

Cadmium (Cd) is a human carcinogen that likely acts via epigenetic mechanisms. However, the precise role of Cd in melanoma remains to be defined. The goals of this study are to: (i) examine the effect of Cd on the proliferation rate of cutaneous and uveal melanoma cells; (ii) identify the genes affected by Cd exposure; (iii) understand whether epigenetic changes are involved in the response to Cd. The cell growth capacity increased at 48 h after Cd treatment at doses ranging from 0.5 to 10 µM. The research on the key genes regulating proliferation has shown that aberrant methylation is responsible for silencing of p16(INK4A) and caspase 8 in uveal and cutaneous melanoma cells, respectively. The methylation and expression patterns of p14(ARF), death receptors 4/5, and E-cadherin remained unmodified after Cd treatment in all the cell lines analyzed. Ectopic expression of p16(INK4A) abolished the overgrowth of uveal melanoma cells in response to Cd and the overexpression of caspase 8 drastically increased the apoptotic rate of Cd-treated cutaneous melanoma cells. In conclusion, our data suggest that hypermethylation of p16(INK4A) and caspase 8 represents the most common event linked to Cd-induced stimulation of cell growth and inhibition of cell death pathway in melanoma.


Asunto(s)
Cloruro de Cadmio/toxicidad , Epigénesis Genética/efectos de los fármacos , Melanoma/inducido químicamente , Caspasa 8/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Neoplasias Cutáneas/inducido químicamente , Proteína p14ARF Supresora de Tumor/efectos de los fármacos , Neoplasias de la Úvea/inducido químicamente
9.
Acta Oncol ; 47(6): 1062-70, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17934890

RESUMEN

BACKGROUND: The active components of Cannabis sativa L., Cannabinoids, traditionally used in the field of cancer for alleviation of pain, nausea, wasting and improvement of well-being have received renewed interest in recent years due to their diverse pharmacologic activities such as cell growth inhibition, anti-inflammatory activity and induction of tumor regression. Here we used several experimental approaches, which identified delta-9-tetrahydrocannabinol (Delta(9)-THC) as an essential mediator of cannabinoid antitumoral action. METHODS AND RESULTS: Administration of Delta(9)-THC to glioblastoma multiforme (GBM) cell lines results in a significant decrease in cell viability. Cell cycle analysis showed G(0/1) arrest and did not reveal occurrence of apoptosis in the absence of any sub-G(1) populations. Western blot analyses revealed a THC altered cellular content of proteins that regulate cell progression through the cell cycle. The cell content of E2F1 and Cyclin A, two proteins that promote cell cycle progression, were suppressed in both U251-MG and U87-MG human glioblastoma cell lines, whereas the level of p16(INK4A), a cell cycle inhibitor was upregulated. Transcription of thymidylate synthase (TS) mRNA, which is promoted by E2F1, also declined as evident by QRT-PCR. The decrease in E2F1 levels resulted from proteasome mediated degradation and was prevented by proteasome inhibitors. CONCLUSIONS: Delta(9)-THC is shown to significantly affect viability of GBM cells via a mechanism that appears to elicit G(1) arrest due to downregulation of E2F1 and Cyclin A. Hence, it is suggested that Delta(9)-THC and other cannabinoids be implemented in future clinical evaluation as a therapeutic modality for brain tumors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Dronabinol/farmacología , Factor de Transcripción E2F1/efectos de los fármacos , Factor de Transcripción E2F1/metabolismo , Glioblastoma/tratamiento farmacológico , Western Blotting , Neoplasias Encefálicas/metabolismo , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Factor de Transcripción E2F1/genética , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/metabolismo , Humanos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timidilato Sintasa/efectos de los fármacos , Timidilato Sintasa/metabolismo , Factores de Tiempo , Regulación hacia Arriba
10.
Brain Res ; 1188: 25-34, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18048012

RESUMEN

Although meningiomas represent the most common class of tumors of the central nervous system, the molecular events underlying their genesis and development are still not well defined, and therapeutic approaches based on the genetics of these tumors are currently lacking. In the present study we have used the immunoblotting technique to show that the p16(INK4A), Cdk6 and pRB proteins are differentially expressed in primary meningioma cells with 20-, 30- and 36-fold difference between the lowest and the highest levels of each protein, respectively. In addition, we present evidence that the level of the anti-apoptosis survivin protein is high in these benign tumors. Moreover, the annexin V-associated flow cytometry technique was used to show that 60% of meningioma cell cultures underwent apoptosis in response to both gamma-rays and cisplatin, and 50% of these cells exhibited significant sensitivity to hydroxyurea. These agents triggered apoptosis through the mitochondrial pathway, by increasing the Bax/Bcl-2 ratio. Interestingly, the induction of apoptosis following radiation and cisplatin was significant in all cells that expressed low levels of p16(INK4A), Cdk6 and pRB proteins. These data shed more light on the molecular biology of meningioma cells and suggest that survivin and proteins of the RB pathway could play a determinant role in the development and the treatment of meningiomas.


Asunto(s)
Apoptosis/fisiología , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína de Retinoblastoma/metabolismo , Adolescente , Adulto , Anciano , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Quinasa 6 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de la radiación , Femenino , Citometría de Flujo , Humanos , Hidroxiurea/farmacología , Immunoblotting , Proteínas Inhibidoras de la Apoptosis , Masculino , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/radioterapia , Meningioma/tratamiento farmacológico , Meningioma/radioterapia , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/efectos de la radiación , Persona de Mediana Edad , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/efectos de la radiación , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de la radiación , Radioterapia , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Survivin , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Regulación hacia Arriba/efectos de la radiación , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/efectos de la radiación
11.
Toxicol Appl Pharmacol ; 225(3): 293-9, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17920094

RESUMEN

Drug-induced liver injury, including drug-induced hepatotoxicity during the treatment of tuberculosis infection, is a major health problem with increasingly significant challenges to modern hepatology. Therefore, the assessment and monitoring of the hepatotoxicity of antituberculosis drugs for prevention of liver injury are great concerns during disease treatment. The recently emerged data showing the ability of toxicants, including pharmaceutical agents, to alter cellular epigenetic status, open a unique opportunity for early detection of drug hepatotoxicity. Here we report that treatment of male Wistar rats with antituberculosis drug pyrazinamide at doses of 250, 500 or 1000 mg/kg/day body weight for 45 days leads to an early and sustained decrease in cytosine DNA methylation, progressive hypomethylation of long interspersed nucleotide elements (LINE-1), and aberrant promoter hypermethylation of placental form glutathione-S-transferase (GSTP) and p16(INK4A) genes in livers of pyrazinamide-treated rats, while serum levels of bilirubin and activity of aminotransferases changed modestly. The early occurrence of these epigenetic alterations and their association with progression of liver injury specific pathological changes indicate that alterations in DNA methylation may be useful predictive markers for the assessment of drug hepatotoxicity.


Asunto(s)
Antituberculosos/toxicidad , Epigénesis Genética/efectos de los fármacos , Hígado/efectos de los fármacos , Pirazinamida/toxicidad , Animales , Antituberculosos/administración & dosificación , Bilirrubina/sangre , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Citosina/metabolismo , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Gutatión-S-Transferasa pi/efectos de los fármacos , Gutatión-S-Transferasa pi/genética , Hígado/patología , Elementos de Nucleótido Esparcido Largo/efectos de los fármacos , Elementos de Nucleótido Esparcido Largo/genética , Masculino , Pirazinamida/administración & dosificación , Distribución Aleatoria , Ratas , Ratas Wistar , Transaminasas/efectos de los fármacos , Transaminasas/metabolismo
12.
J Gerontol A Biol Sci Med Sci ; 62(3): 246-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17389721

RESUMEN

DNA demethylating agents are used to reverse epigenetic silencing of tumor suppressors in cancer therapeutics. Understanding of the molecular and cellular factors involved in DNA demethylation-induced gene desilencing and senescence is still limited. We have tested the involvement of two stress chaperones, Pex19p and mortalin, in 5-Aza-2' deoxycytidine (5AZA-dC; DNA demethylating agent)-induced senescence. We found that the cells overexpressing these chaperones were highly sensitive to 5AZA-dC, and their partial silencing eliminated 5AZA-dC-induced senescence in human osteosarcoma cells. We demonstrate that these chaperones modulate the demethylation and chromatin remodeling-dependent (as accessed by p16(INK4A) expression) and remodeling-independent (such as activation of tumor suppressor p53 pathway) senescence response of cells. Furthermore, we found the direct interactions of 5AZA-dC with these chaperones that may alter their functions. We conclude that both mortalin and Pex19p are important mediators, prognostic indicators, and tailoring tools for 5AZA-dC-induced senescence in cancer cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Senescencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/farmacología , Proteínas de Choque Térmico/farmacología , Lipoproteínas/farmacología , Proteínas de la Membrana/farmacología , Chaperonas Moleculares/farmacología , Osteosarcoma/patología , Azacitidina/farmacología , Biomarcadores/análisis , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Decitabina , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen/efectos de los fármacos , Genes p53/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas de Choque Térmico/genética , Humanos , Lipoproteínas/genética , Proteínas de la Membrana/genética , Chaperonas Moleculares/genética , Proteínas Supresoras de Tumor/efectos de los fármacos
13.
Mol Cancer Ther ; 4(7): 1105-13, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16020668

RESUMEN

We discovered a series of salicylhydrazide class of compounds with remarkable anticancer activity against a panel of hormone receptor-positive and -negative cell lines. In the present study, we evaluated the in vitro activity of SC21 and SC23 against a range of human tumor cell types and the in vivo efficacy of compound SC21 in a PC3 human prostate cancer xenograft model in mice. We also determined the effects of SC21 on cell cycle regulation and apoptosis. Our in vitro results show that salicylhydrazides are highly potent compounds effective in both hormone receptor-positive and -negative cancer cells. SC21 induced apoptosis and blocked the cell cycle in G(0)/G(1) or S phase, depending on the cell lines used and irrespective of p53, p21, pRb, and p16 status. SC21 effectively reduced the tumor growth in mice without apparent toxicity. Although the mechanism of action of SC21 is not completely elucidated, the effect on cell cycle, the induction of apoptosis and the activity against a panel of tumor cell lines of different origins prompted us to carry out an in-depth preclinical evaluation of SC21.


Asunto(s)
Antineoplásicos/farmacología , Hidrazinas/farmacología , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Salicilamidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Cisplatino/farmacología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/efectos de los fármacos , Receptores de Estrógenos/efectos de los fármacos , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Neurosurg ; 102(1): 98-108, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15658102

RESUMEN

OBJECT: Brain tumors, including gliomas, develop several months after rats are exposed in utero to N-ethyl-N-nitroso-urea (ENU). Although pathological changes cannot be detected until these animals are several weeks old, the process that eventually leads to glioma formation must begin soon after exposure given the rapid clearance of the carcinogen and the observation that transformation of brain cells isolated soon after exposure occasionally occurs. This model can therefore potentially provide useful insights about the early events that precede overt glioma formation. The authors hypothesized that future glioma cells arise from stem/progenitor cells residing in or near the subventricular zone (SVZ) of the brain. METHODS: Cells obtained from the SVZ or corpus striatum in ENU-exposed and control rats were cultured in an epidermal growth factor (EGF)-containing, chemically defined medium. Usually, rat SVZ cells cultured in this manner (neurospheres) are nestin-positive, undifferentiated, and EGF-dependent and undergo cell senescence. Consistent with these prior observations, control SVZ cells undergo senescence by the 12th to 15th doubling (20 of 20 cultures). In contrast, three of 15 cultures of cells derived from the SVZs of individual ENU-treated rats continue to proliferate for more than 60 cell passages. Each of these nestin-expressing immortalized cell lines harbored a common homozygous deletion spanning the INK4a/ARF locus and was unable to differentiate into neural lineages after exposure to specific in vitro stimuli. Nevertheless, unlike the rat C6 glioma cell line, these immortalized cell lines demonstrate EGF dependence and low clonogenicity in soft agar and did not form tumors after intracranial transplantation. CONCLUSIONS: Data in this study indicated that immortalized cells may represent glioma precursors that reside in the area of the SVZ after ENU exposure that may serve as a reservoir for further genetic and epigenetic hits that could eventually result in a full glioma phenotype.


Asunto(s)
Factores de Ribosilacion-ADP/efectos de los fármacos , Factores de Ribosilacion-ADP/deficiencia , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Etilnitrosourea/toxicidad , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Animales , Encéfalo/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Diferenciación Celular/fisiología , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Cartilla de ADN/genética , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Masculino , Reacción en Cadena de la Polimerasa , Embarazo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Proteína p14ARF Supresora de Tumor/genética
15.
Cancer Lett ; 217(1): 33-41, 2005 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-15596294

RESUMEN

All-trans-retinoic acid (atRA) exerts its effects via apoptosis and cell cycle re-distribution. However, the mechanisms behind the effects have not been fully understood. In this study, we used a model system of matched primary and metastatic melanoma cells to investigate whether expression of Id1 and p16 proteins were involved in atRA-induced apoptosis and cell cycle re-distribution. Melanoma cells were exposed to 0.1 or 10 microM atRA for 1-96 h. Apoptosis and cell cycle were measured by flow cytometry. Expression of Id1 and p16 proteins was examined by Western blotting and immunocytochemistry. After exposure to atRA we found a marked increase in apoptosis and cell cycle re-distribution in both primary and metastatic melanoma cells. Expression level of Id1 protein was decreased and the p16 was increased in a dose- and time-dependent (P<0.05) manner after treatment with atRA. Alterations of these proteins were more pronounced in the primary melanoma cells than the matched metastases (P<0.05). These data suggested that the alterations of Id1 and/or p16 proteins were involved in atRA-induced apoptosis and cell cycle re-distribution in melanoma. These expression profiles of Id1 and p16 proteins may provide molecular evidence for better chemotherapy primarily for early stages of melanoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Melanoma/metabolismo , Proteínas Represoras/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Tretinoina/farmacología , Apoptosis/fisiología , Western Blotting , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Técnicas In Vitro , Proteína 1 Inhibidora de la Diferenciación , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/efectos de los fármacos , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 35(6): 792-6, 2004 Nov.
Artículo en Chino | MEDLINE | ID: mdl-15573756

RESUMEN

OBJECTIVE: To inquire into the mechanism of CDP (a purified component of Chinese crude drug) in inhibiting the breast cancer T47D cell growth. METHODS: T47D cells were treated with different concentration of 5-azacytidine (5-aza-CR) and CDP for several days. The growth rate was assessed by cell proliferation experiment (MTT colorimetric assay). The changes in apoptic peak and cell cycle distribution were detected by flow cytometry (FCM). The levels of methylation and unmethylation status of p16 were detected by methylation specific PCR (MSP). RESULTS: After treatment with the two drugs, the cell growth rate decreased in a dose and time dependent manner (P<0.05). The cell cycle was influenced by the well-chosen concentration of 5-aza-CR (2 micromol/L) and CDP (50 micromol/L): the cell number increased from 65.1% to 71.3%, 84.3% in G0/G1 phase and decreased from 19.4% to 14.3%, 7.2% in S phase. Demethylation on p16 gene occurred after treatment with any of the two drugs for 6 days. CONCLUSION: CDP can reverse p16 hypermethylation and may hence inhibit the proliferation of tumor cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/genética , Metilación de ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Genes p16/efectos de los fármacos , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Metilasas de Modificación del ADN/antagonistas & inhibidores , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Humanos
17.
Oncogene ; 23(17): 3116-21, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-14743210

RESUMEN

Exogenous expression of the catalytic subunit of telomerase, hTERT, in a normal human foreskin fibroblast cell strain resulted in telomerase activity and an extended proliferative lifespan prior to a period of crisis. Three immortalized cell lines with stably maintained telomere lengths were established from cells that escaped crisis. Each of these cultures underwent a significant downregulation of p16(INK4a) expression due to gene deletion events. One cell line also acquired mutations in both alleles of the p53 tumor suppressor gene. Downregulation of p16(INK4a) and loss of wild-type p53 expression occurred after escape from crisis, so these mutations are most likely not required for immortalization of these cells but rather were selected for during continuous growth in vitro. These findings emphasize the need for caution in the use of hTERT-immortalized cells in studies of normal cell biology or in tissue engineering and the need to monitor for genetic instability and the accumulation of mutations in both the p16(INK4a)/pRb and p53 pathways.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Genes p53 , Telomerasa/genética , Línea Celular Transformada , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Proteínas de Unión al ADN , Dactinomicina/farmacología , Fibroblastos , Humanos , Recién Nacido , Cariotipificación , Masculino , Mutación , Telomerasa/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
18.
Mol Cell Biochem ; 247(1-2): 55-60, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12841631

RESUMEN

The importance of bone marrow mesenchymal stem cells in hemopoiesis has been definitely demonstrated. Thus, their impairment might cause profound alteration on production and maturation of blood cells. In the present paper, we investigated, for the first time, the effect of retinoic acid, an important antileukemic molecule, on the proliferation of primary cultures of human bone marrow mesenchymal stem cells. We demonstrated that retinoic acid, at a pharmacological concentration, hampers strongly the growth of the cells, without inducing osteoblastic differentiation. The analysis of cell division cycle machinery showed that the antiproliferative effect is associated with (i) the up-regulation of two cyclin-dependent kinase inhibitors, namely p27Kip1 and p16INK4A, and (ii) the down-regulation of cyclin-dependent kinase 2 activity and pRB phosphorylation. The reported findings represent novel insights into the antileukemic effects of the drug and contribute in clarifying the molecular mechanism of its pharmacological activity.


Asunto(s)
Células de la Médula Ósea/citología , Proteínas de Ciclo Celular/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Mesodermo/citología , Células Madre/metabolismo , Tretinoina/farmacología , Proteínas Supresoras de Tumor/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Quinasas CDC2-CDC28/efectos de los fármacos , Quinasas CDC2-CDC28/metabolismo , Proteínas de Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Quinasa 2 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Humanos , Mesodermo/efectos de los fármacos , Fenotipo , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
19.
Melanoma Res ; 13(3): 231-8, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12777976

RESUMEN

Flavopiridol is the first cyclin-dependent kinase inhibitor to enter clinical trials. Flavopiridol has been shown to mimic, in part, the effect of the cell cycle control gene p16, which is frequently lost or mutated in malignant melanoma, making it an ideal candidate for targeted therapy in this disease. In these studies we investigated the effect of flavopiridol, at various concentrations, on the growth and gene expression of nine human melanoma cell lines with intact, absent or mutated p16. A cytostatic effect of flavopiridol on the growth of six melanoma cell lines with a mutated or non-expressed p16 (p16-) was seen at low concentrations of flavopiridol (mean 50% inhibitory concentration [IC(50)] = 12.5 nM), while the three melanoma cell lines with intact p16 (p16+) required higher concentrations (mean IC(50) = 25 nM) to produce this effect. Apoptotic cell death increased with increasing concentrations of flavopiridol in both p16- and p16+ cells. Exposure of cells to high flavopiridol concentrations (>100 nM) resulted in decreased expression of genes downstream in the normal p16 cell cycle control pathway (Rb and E2F) and the anti-apoptotic gene BCL2. No change in BCL2 expression was found after exposure to IC(50) concentrations of flavopiridol. These data indicate that flavopiridol in low, clinically achievable concentrations may have significant cytostatic effects, particularly in p16- melanoma cells, and may provide new molecular-based therapies for melanoma, particularly when combined with agents that target anti-apoptotic mechanisms.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Flavonoides/farmacología , Genes p16 , Melanoma/genética , Melanoma/patología , Piperidinas/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas/citología
20.
Mol Carcinog ; 36(3): 153-60, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12619038

RESUMEN

The neurotrophin (NTR) receptor (p75(NTR)) is a cell-surface glycoprotein that binds to the neurotrophin family of growth factors, of which the prototypic member is nerve growth factor (NGF). This receptor was previously shown to retard cell-cycle progression by inducing accumulation of cells in G(1) with a concomitant reduction of cells in the S phase of the cell cycle. Furthermore, p75(NTR) was shown to be an effective tumor suppressor of bladder cancer cell growth in vivo. In order to investigate the mechanism of p75(NTR)-dependent suppression of cell-cycle progression, we utilized transgenic clones of bladder tumor cells that express p75(NTR) in increasing concentrations to demonstrate an effect of p75(NTR) on the levels of cell-cycle regulatory proteins that modulate proliferation of tumor cells. A rank-order (dose-dependent) increase in p75(NTR) protein expression was associated with a decrease in cell proliferation. This p75(NTR)-dependent suppression of proliferation was rescued with NGF. In the absence of ligand, a dose-dependent increase in p75(NTR) protein expression was associated with reduced expression of cyclin D1, cyclin E, and cyclin-dependent kinase 2 (cdk2) as well as decreased cdk2 activity. There was also a decrease in the expression of hyper-phosphorylated retinoblastoma protein, the transcription factor E2F1, and proliferating cell nuclear antigen, and there was an increase in expression of hypophosphorylated Rb and the cdk inhibitor p16(Ink4a) with increasing p75(NTR) expression. Treatment of tumor cells with NGF ameliorated these p75(NTR)-dependent changes in the levels of cell-cycle regulatory proteins and rescued the tumor cells from p75(NTR)-dependent inhibition of proliferation. Hence, it can be concluded that p75(NTR) inhibits proliferation by altering the expression of cell-cycle regulatory proteins and that NGF ameliorates this effect.


Asunto(s)
Quinasas CDC2-CDC28 , Proteínas de Ciclo Celular , Ciclo Celular/fisiología , Proteínas de Unión al ADN , Receptores de Factor de Crecimiento Nervioso/genética , Neoplasias de la Vejiga Urinaria/patología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Células Epiteliales/patología , Genes Supresores de Tumor , Factor de Crecimiento Nervioso/farmacología , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor de Factor de Crecimiento Nervioso , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptores de Factor de Crecimiento Nervioso/metabolismo , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...