Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 117(8): 1154-1163, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-28873083

RESUMEN

BACKGROUND: Overexpression of epidermal growth factor receptor (EGFR) occurs in approximately 90% of head and neck squamous cell carcinoma (HNSCC), and is correlated with poor prognosis. Thus, targeting EGFR is a promising strategy for treatment of HNSCC. Several small molecule EGFR inhibitors have been tested in clinical trials for treatment of HNSCC, but none of them are more effective than the current chemotherapeutic drugs. Thus, it is urgently needed to develop novel EGFR inhibitors for HNSCC treatment. METHODS: By screening an in-house focused library containing approximately 650 000 known kinase inhibitors and kinase inhibitor-like compounds containing common kinase inhibitor core scaffolds, we identified SKLB188 as a lead compound for inhibition of EGFR. The anticancer effects of SKLB188 on HNSCC cells were investigated by in vitro cell growth, cell cycle and apoptosis assays, as well as in vivo FaDu xenograft mouse model. Molecular docking, in vitro kinase profiling and western blotting were performed to characterise EGFR as the molecular target. RESULTS: SKLB188 inhibited HNSCC cell proliferation by inducing G1 cell cycle arrest, which was associated with downregulating the expression of Cdc25A, cyclins D1/A and cyclin-dependent kinases (CDK2/4), and upregulating the expression of cyclin-dependent kinase (CDK) inhibitors (p21Cip1 and p27Kip1), leading to decreased phosphorylation of Rb. SKLB188 also induced caspase-dependent apoptosis of HNSCC cells by downregulating the expression of Mcl-1 and survivin. Molecular docking revealed that SKLB188 could bind to the kinase domain of EGFR through hydrogen bonds and hydrophobic interactions. In vitro kinase assay showed that SKLB188 inhibited the activity of a recombinant human EGFR very potently (IC50=5 nM). Western blot analysis demonstrated that SKLB188 inhibited the phosphorylation of EGFR and its downstream targets, extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) and Akt in the cells. In addition, SKLB188 dose-dependently inhibited FaDu xenograft growth in nude mice, and concurrently inhibited the phosphorylation of Erk1/2 and Akt in the tumours. CONCLUSIONS: SKLB188 potently inhibits the growth of HNSCC cells in vitro and in vivo by targeting EGFR signalling. The results provide a basis for further clinical investigation of SKLB188 as a targeted therapy for HNSCC. Our findings may open a new avenue for development of novel EGFR inhibitors for treatment of HNSCC and other cancers.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Purinas/farmacología , Animales , Western Blotting , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/efectos de los fármacos , Fosfatasas cdc25/metabolismo
2.
Cell Biol Int ; 39(9): 1044-52, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25857830

RESUMEN

The purpose of the study was to examine mechanisms controlling cell cycle progression/arrest and differentiation of mouse C2C12 myoblasts exposed to long-chain saturated fatty acid salt, palmitate. Treatment of proliferating myoblasts with palmitate (0.1 mmol/l) markedly decreased myoblast number. Cyclin A and cyclin D1 levels decreased, whereas total p21 and p21 complexed with cyclin-dependent kinase-4 (cdk4) increased in myoblasts treated with palmitate. In cells induced to differentiation addition of palmitate augmented the level of cyclin D3, the early (myogenin) and late (α-actinin, myosin heavy chain) markers of myogenesis, and caused an increase of myotube diameter. In conclusion, exposure to palmitate inhibits proliferation of myoblasts through a decrease in cyclin A and cyclin D1 levels and an increase of p21-cdk4 complex formation; however, it promotes cell cycle exit, myogenic differentiation and myotube growth.


Asunto(s)
Mioblastos Esqueléticos/efectos de los fármacos , Palmitatos/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Ciclina A/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Miogenina/efectos de los fármacos
3.
Postepy Hig Med Dosw (Online) ; 69: 1259-67, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26671917

RESUMEN

BACKGROUND: Arsenic trioxide (ATO) is an effective drug used in acute promyelocytic leukemia (AML). Many reports suggest that ATO can also be applied as an anticancer agent for solid tumors in the future. The influence of arsenic trioxide on the expression of different cell cycle regulators is poorly recognized. The purpose of the current study is to investigate how arsenic trioxide affects cyclin A expression and localization in the A549 cell line. MATERIALS AND METHODS: Morphological and ultrastructural changes in A549 cells were observed using light and transmission electron microscopes. Cyclin A localization was determined by immunofluorescence. Image-based cytometry was applied to evaluate the effect of arsenic trioxide on apoptosis and the cell cycle. Expression of cyclin A mRNA was quantified by real-time PCR. RESULTS: After treatment with arsenic trioxide, increased numbers of cells with cytoplasmic localization of cyclin A were observed. The doses of 10 and 15 µM ATO slightly reduced expression of cyclin A mRNA. The apoptotic phenotype of cells was poorly represented, and the Tali imagebased cytometry analysis showed low percentages of apoptotic cells. The A549 population displayed an enriched fraction of cells in G0/G1 phase in the presence of 5µM ATO, whereas starting from the higher concentrations of the drug, i.e. 10 and 15 µM ATO, the G2/M fraction was on the increase. DISCUSSION: Low expression of cyclin A in the A549 cell line may constitute a potential factor determining arsenic trioxide resistance. It could be hypothesized that the observed alterations in cyclin A expression/distribution may correlate well with changes in cell cycle regulation in our model, which in turn determines the outcome of the treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Ciclo Celular/efectos de los fármacos , Ciclina A/genética , Óxidos/farmacología , Trióxido de Arsénico , Línea Celular , Ciclina A/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Citometría de Imagen , Microscopía Electrónica
4.
J Exp Ther Oncol ; 9(3): 175-81, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22070048

RESUMEN

Ovarian cancer is the leading cause of death from gynecological cancer. The high mortality rate reflets the lack of early diagnosis and limited treatment alternatives. We have observed a number of properties of zinc cytotoxicity that make it attractive from a therapeutic standpoint. Using SKOV3 and ES2 cells, ovarian cancer cell lines that demonstrate varied degrees of resistance to known therapeutics, we show that zinc killing is time and concentration dependent. Death is preceded by distinct changes in cell shape and size. The effects of zinc are additive with cisplatin or doxorubicin, whose morphological effects are distinct from those of zinc. Cytotoxicity of paclitaxel is minimal, making it difficult to determine additivity with zinc. Paclitaxel results in changes in cell shape and size similar to those of zinc but has different effects on cell cycle progression and cyclin expression. The data indicate that the means by which zinc kills ovarian cancer cells is distinct from currently used chemotherapeutics. Based on the properties reported here, zinc has the potential to be developed as either a primary treatment or as a second line of defense against cancers that have developed resistance to currently used chemotherapeutics.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Oligoelementos/farmacología , Zinc/farmacología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D/efectos de los fármacos , Ciclina D/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Paclitaxel/farmacología , Piridinas/farmacología , Tionas/farmacología
5.
Theranostics ; 11(20): 10047-10063, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34815803

RESUMEN

Parkin (PK) is an E3-ligase harboring tumor suppressor properties that has been associated to various cancer types including glioblastoma (GBM). However, PK is also a transcription factor (TF), the contribution of which to GBM etiology remains to be established. Methods: The impact of PK on GBM cells proliferation was analyzed by real-time impedance measurement and flow cytometry. Cyclins A and B proteins, promoter activities and mRNA levels were measured by western blot, luciferase assay and quantitative real-time PCR. Protein-protein and protein-promoter interactions were performed by co-immunoprecipitation and by ChIP approaches. The contribution of endogenous PK to tumor progression in vivo was performed by allografts of GL261 GBM cells in wild-type and PK knockout mice. Results: We show that overexpressed and endogenous PK control GBM cells proliferation by modulating the S and G2/M phases of the cell cycle via the trans-repression of cyclin A and cyclin B genes. We establish that cyclin B is regulated by both E3-ligase and TF PK functions while cyclin A is exclusively regulated by PK TF function. PK invalidation leads to enhanced tumor progression in immunocompetent mice suggesting an impact of PK-dependent tumor environment to tumor development. We show that PK is secreted by neuronal cells and recaptured by tumor cells. Recaptured PK lowered cyclins levels and decreased GBM cells proliferation. Further, PK expression is decreased in human GBM biopsies and its expression is inversely correlated to both cyclins A and B expressions. Conclusion: Our work demonstrates that PK tumor suppressor function contributes to the control of tumor by its cellular environment. It also shows a key role of PK TF function in GBM development via the control of cyclins in vitro and in vivo. It suggests that therapeutic strategies aimed at controlling PK shuttling to the nucleus may prove useful to treat GBM.


Asunto(s)
Glioblastoma/terapia , Ubiquitina-Proteína Ligasas/uso terapéutico , Animales , Neoplasias Encefálicas/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclinas/genética , Femenino , Citometría de Flujo/métodos , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor/fisiología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Modelos Animales , ARN Mensajero
6.
J Cell Physiol ; 223(2): 327-34, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20054826

RESUMEN

Cdc20, an activator of the anaphase promoting complex/cyclosome (APC/C) ubiquitin ligase, initiates the destruction of key mitotic regulators to facilitate mitosis, while it is negatively regulated by the spindle assembly checkpoint (SAC) to prevent premature anaphase entry. Activation of the p38 mitogen-activated protein kinase could contribute to mitotic arrest, but the underlying mechanism is unknown. Here we report a novel pathway in which the p38 signaling triggers Cdc20 destruction under SAC elicited by cadmium, a human carcinogen. We found that the cadmium-induced prometaphase arrest was linked to decreased Cdc20 and accumulated cyclin A protein levels in human cells, whereas the activity of cyclin B1-Cdk1 was unaffected. The Cdc20 half-life was markedly shortened along with its ubiquitination and degradation via 26S proteasome in cadmium-treated asynchronous or G(2)-enriched cells. Depletion of APC3 markedly suppressed the cadmium-induced Cdc20 ubiquitination and proteolysis, while depletion of Cdh1, another activator of APC/C, did not. Intriguingly, blockage of p38 activity restored the Cdc20 levels for continuing mitosis under cadmium, while inhibition of JNK activity had no effect. The cadmium-induced Cdc20 proteolysis was also suppressed during transient depletion of p38alpha or stable expression a dominant negative form of p38. Inhibition of p38 abolished the induction of Mad2-Cdc20-APC3 complex by cadmium. Moreover, forced expression of MKK6-p38 signaling could promote Cdc20 degradation in a Cdh1-independent APC/C pathway. In summary, accelerated ubiquitination and proteolysis of Cdc20 is essential for prometaphase arrest that is mediated via the p38 signaling during SAC activation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Genes cdc/fisiología , Huso Acromático/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Antígenos CD , Subunidad Apc3 del Ciclosoma-Complejo Promotor de la Anafase , Cadherinas/metabolismo , Cadmio/farmacología , Proteínas Cdc20 , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Inhibidores Enzimáticos/farmacología , Genes cdc/efectos de los fármacos , Humanos , MAP Quinasa Quinasa 6/efectos de los fármacos , MAP Quinasa Quinasa 6/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Mitógenos/farmacología , Péptido Hidrolasas/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Prometafase/efectos de los fármacos , Prometafase/fisiología , Huso Acromático/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Ubiquitinación/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
7.
Amino Acids ; 36(2): 273-82, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18368465

RESUMEN

Polyamines are important for cell growth and proliferation and they are formed from arginine and ornithine via arginase and ornithine decarboxylase (ODC). Arginine may alternatively be metabolised to NO via NO synthase. Here we study if vascular smooth muscle cell proliferation can be reversed by polyamine synthesis inhibitors and investigate their mechanism of action. Cell proliferation was assessed in cultured vascular smooth muscle A7r5 cells and in endothelium-denuded rat arterial rings by measuring [3H]-thymidine incorporation and by cell counting. Cell cycle phase distribution was determined by flow cytometry and polyamines by HPLC. Protein expression was determined by Western blotting. The ODC inhibitor DFMO (1-10 mM) reduced polyamine concentration and attenuated proliferation in A7r5 cells and rat tail artery. DFMO accumulated cells in S phase of the cell cycle and reduced cyclin A expression. DFMO had no effect on cell viability and apoptosis as assessed by fluorescence microscopy. Polyamine concentration and cellular proliferation were not affected by the arginase inhibitor NOHA (100-200 microM) and the NO synthase inhibitor L-NAME (100 microM). Lack of effect of NOHA was reflected by absence of arginase expression. Polyamine synthesis inhibition attenuates vascular smooth muscle cell proliferation by reducing DNA synthesis and accumulation of cells in S phase, and may be a useful approach to prevent vascular smooth muscle cell proliferation in cardiovascular diseases.


Asunto(s)
Músculo Liso Vascular/citología , Poliaminas/antagonistas & inhibidores , Fase S , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Ornitina Descarboxilasa/metabolismo , Inhibidores de la Ornitina Descarboxilasa , Poliaminas/metabolismo , Ratas , Ratas Sprague-Dawley , Fase S/efectos de los fármacos
8.
Anticancer Drugs ; 20(4): 230-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19288605

RESUMEN

A number of polyamine analogs are currently used in various clinical trials as cancer treatment and it is important to investigate their effects not only on cancer cells but also on normal cells. Treatment with polyamine analogs depletes cells of polyamines and inhibits cell proliferation, but the analogs cannot take over the normal function of the natural polyamines in the cell. In this study, the normal-like breast epithelial cell line MCF-10A was treated with the polyamine analog N',N"-diethylnorspermine (DENSPM). The cells were then studied using a bromodeoxyuridine- DNA flow cytometry method as well as western blot. The ability of both normal-like and breast cancer cells to recover from DENSPM treatment was also studied. DENSPM treatment of MCF-10A cells resulted in a prolongation of the S and G2 +M phases, followed by a G1/S block. The p53/p21/RB1 pathway was involved in the G1/S block as shown by increased levels of p53 and p21 detected by western blot. Decreased levels of cyclin E1, cyclin A2, and cyclin B1 in DENSPM-treated cells can explain the prolongation of cell cycle phases that occurred before the G1/S block. We also show that MCF-10A cells rapidly recover from DENSPM-induced growth inhibition in contrast to four human breast cancer cell lines. The goal of cancer treatment is to cause minimal and reversible damage to normal cells, while cancer cells should be eliminated. Altogether, the data show that treatment with polyamine analogs spares normal cells, while negatively affecting the cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Espermina/análogos & derivados , Western Blotting , Neoplasias de la Mama/patología , Bromodesoxiuridina , Línea Celular , Línea Celular Tumoral , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina A2 , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , ADN , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Espermina/farmacología
9.
Int J Cancer ; 123(12): 2931-8, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18798266

RESUMEN

OSU03012 is a non-COX inhibiting celecoxib derivative with growth inhibiting and apoptotic activity in many cancer cell lines. To investigate mechanisms related to cell cycle proteins in growth inhibition and apoptosis induced by OSU03012, the primary human oral epithelial cell line, TE1177, was transformed with HPV16 E6 (TE/E6), HPV16 E7 (TE/E7) or empty vector (TE/V). TE/E6 cell lines exhibiting low levels of p53 and undetectable levels of p21(WAF1/CIP1) were sensitized to the growth inhibiting and apoptotic effects of OSU03012. The TE/E7 cell lines expressing low levels of Rb and elevated levels of p53 and p21(WAF1/CIP1) were resistant. OSU03012 reduced the number of cells in the S phase of the TE/E7 and TE/V cell lines with intact p53-p21(WAF1/CIP1) checkpoint, but not in the checkpoint defective TE/E6 cell lines. Treatment with OSU03012 also markedly reduced the levels of cyclin A and Cdk2 in TE/E7 and TE/V, but not in TE/E6 cell lines, which had significantly enhanced basal levels of cyclin A and Cdk2. Consistent with the TE/E6 cell line, p21(WAF1/CIP1)-/- mouse embryo fibroblasts were more sensitive to OSU03012-induced apoptosis as evidenced by PARP and caspase 3 cleavages. These data suggest that p21(WAF1/CIP1) is an important factor in the sensitivity of cells to the growth inhibiting and apoptotic effects of OSU03012.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Mucosa Bucal/efectos de los fármacos , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Bromodesoxiuridina/metabolismo , Línea Celular , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Embrión de Mamíferos , Fibroblastos/efectos de los fármacos , Citometría de Flujo , Fase G1/efectos de los fármacos , Silenciador del Gen , Humanos , Inmunoprecipitación , Ratones , Mucosa Bucal/citología , Mucosa Bucal/virología , Proteínas Oncogénicas Virales/efectos de los fármacos , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/metabolismo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Infecciones Tumorales por Virus/metabolismo
10.
Int J Oncol ; 33(3): 613-21, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18695893

RESUMEN

In mammalian cells, the H2AX histone is rapidly phosphorylated upon the induction of DNA double strand breaks and promotes their repair, which is required for preserving genomic integrity. Etoposide is an inhibitor of DNA topoisomerase II, which causes DNA breaks and induces H2AX phosphorylation. To elucidate whether H2AX may affect cellular sensitivity to etoposide, we studied the response to this agent in immortalized embryonic fibroblasts derived from H2AX knockout mice. Clonogenic assays in cells treated with the drug revealed a greater sensitivity of H2AX null cells compared to wild-type cells, possibly due to the persistence of a higher number of DNA breaks, as detected with the comet assay. In both cell lines, etoposide induced micronuclei formation and nuclear fragmentation; however, in H2AX deficient cells nuclear fragmentation was observed at a lower drug concentration. Flow cytometric analysis showed that etoposide induced a G2/M cell cycle arrest in both cell lines, which occurred at lower drug concentrations in H2AX deficient cells. G2/M arrest was paralleled by an accumulation of cyclin A and cyclin B1, suggesting that treated cells are not able to complete cell cycle correctly and undergo cell death. Taken together, our observations suggest that H2AX takes part to the cellular response to etoposide and confirm its role in the maintenance of genome stability.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos/fisiología , Etopósido/farmacología , Fibroblastos/efectos de los fármacos , Histonas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular , Ensayo Cometa , Ciclina A/biosíntesis , Ciclina A/efectos de los fármacos , Ciclina B/biosíntesis , Ciclina B/efectos de los fármacos , Citometría de Flujo , Histonas/genética , Ratones , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Inhibidores de Topoisomerasa II
11.
Toxicology ; 250(1): 39-46, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18577415

RESUMEN

In the present study, we investigated the effects of manganese chloride (MnCl2) on cell cycle progression in A549 cells used as a model of Mn-induced lung toxicity. Cells were treated with various concentrations of MnCl2 (0, 0.01, 0.1, 0.5, 1.0 or 2.0 mM) for 24, 48 or 72 h. Cell proliferation was determined with MTT assay and mitotic index measurement and apoptosis was measured by flow cytometer. The results showed that MnCl2 inhibited A549 cells proliferation in a dose- and time-dependent manner, and induced apoptosis in A549 cells. When G0/G1 cells obtained by serum starvation were incubated with 0.5 mM of MnCl2 in the presence of 10% serum for several time intervals, the disruption of cell cycle progression was observed. The G0/G1 arrest was induced by MnCl2 treatment at 16 h and the arrest maintained for 8 h. Following the G0/G1 arrest, MnCl2 blocked the cells at S phase at 28 h and the S phase arrest maintained for at least 4 h. And moreover, proteasome inhibitor MG132 was able to prolong the duration of G0/G1 arrest induced by MnCl2 treatment. Results of western blotting assay revealed that cellular Cdk4, Cdk2 and phospho-Cdk2 (Thr160) levels decreased in manganese-treated cells at both 20 and 28 h. In addition, the decreasing of Cyclin A level and the increasing of p53 and WAF1/p21 were also induced by MnCl2 treatment at 20 h. The expression of Cyclin D1, Cyclin E and Cdc25A proteins was not altered in manganese-treated cells at both 20 and 28 h. Our results indicate that MnCl2 orderly induces G0/G1 and S phase arrest in A549 cells, the decreasing of Cdk4, Cdk2 and Cyclin A, and the increasing of p53 and Cdks inhibitor WAF1/p21 might be responsible for the G0/G1 arrest, and the decreasing of Cdk4 and Cdk2 levels for the S phase arrest.


Asunto(s)
Cloruros/toxicidad , Fase G1/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase S/efectos de los fármacos , Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloruros/administración & dosificación , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Compuestos de Manganeso/administración & dosificación , Índice Mitótico , Factores de Tiempo
12.
J Pharm Pharmacol ; 60(11): 1459-63, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18957166

RESUMEN

We reported previously that oroxylin A, a natural product isolated from Scutellariae Radix, was a potent apoptosis inducer of human hepatoma HepG2 cells. In this study, cell-cycle arrest of BGC-823 human gastric carcinoma cells caused by oroxylin A has been investigated. Based on our 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) assay and flow cytometric analysis, treatment of BGC-823 cells with growth suppressive concentrations of oroxylin A caused an irreversible arrest in the G2/M phase of the cell cycle. Western blot analysis demonstrated that oroxylin A-induced cell-cycle arrest in BGC-823 cells was associated with a significant decrease in cdc2/p34, cyclin B1 and cyclin A expression. In addition, oroxylin A-treated cells decreased the expression of Cdk7, which was responsible for the low expression of M phase promoting factor (cyclin B1/Cdc2). The results suggested that oroxylin A induced G2/M phase cell-cycle arrest via inhibiting Cdk7-mediated expression of Cdc2/p34 in human gastric carcinoma BGC-823 cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteína Quinasa CDC2/efectos de los fármacos , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Western Blotting , Proteína Quinasa CDC2/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Citometría de Flujo , Fase G2/efectos de los fármacos , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Sales de Tetrazolio , Tiazoles , Quinasa Activadora de Quinasas Ciclina-Dependientes
13.
Mol Cancer Ther ; 4(7): 1039-46, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16020661

RESUMEN

Cyclin A-associated kinases, such as cyclin-dependent kinase 2 (CDK2), participate in regulating cellular progression from G(1) to S to G(2), and CDK2 has also been implicated in the transition to mitosis. The antitumor properties of CDK inhibitors, alone or in combination with taxanes, are currently being examined in clinical trials. Here, we examined whether the activity of kinases associated with cyclin A (such as CDK2) is important in determining cellular sensitivity to paclitaxel, a taxane and mitotic inhibitor used in chemotherapy for breast and ovarian cancer. We used adenoviral suppression or overexpression to manipulate the expression of CDK2 and cyclin A in one breast cancer and three ovarian cancer cell lines with different sensitivities to paclitaxel and assessed protein expression, kinase activity, cell cycle distribution, and sensitivity to paclitaxel. Transfection of a dominant-negative (DN)-CDK2 evoked resistance to paclitaxel by preventing cellular progression to mitosis through loss of CDK1 activity. Reexpression of wild-type CDK2 in DN-CDK2-transfected cancer cells restored CDK2 activity but not paclitaxel sensitivity. However, expression of cyclin A in DN-CDK2-transfected cells restored their sensitivity to paclitaxel. Although CDK2 activity was not directly involved in paclitaxel sensitivity, cyclin A-associated kinases did up-regulate CDK1 via phosphorylation. We conclude that cyclin A-associated kinase activity is required for these cells to enter mitosis and undergo paclitaxel-induced cell death. Combining taxane chemotherapy with any drug targeting cyclin A-associated kinases (e.g., pure CDK2 inhibitors) should be done with caution, if at all, because of the potential for enhancing taxane resistance.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Ciclina A/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Resistencia a Antineoplásicos , Paclitaxel/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Ciclina A/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/efectos de los fármacos , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Fosforilación , Treonina/metabolismo , Transfección , Células Tumorales Cultivadas
14.
Oncogene ; 18(15): 2461-70, 1999 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10229197

RESUMEN

Oxamflatin [(2E)-5-[3-[(phenylsufonyl) aminol phenyl]-pent-2-en-4-ynohydroxamic acid] induces transcriptional activation of junD and morphological reversion in various NIH3T3-derived transformed cell lines. We found that oxamflatin showed in vitro antiproliferative activity against various mouse and human tumor cell lines with drastic changes in the cell morphology and in vivo antitumor activity against B16 melanoma. Oxamflatin caused an elongated cell shape with filamentous protrusions as well as arrest of the cell cycle at the G1 phase in HeLa cells. These phenotypic changes of HeLa cells were apparently similar to those by trichostatin A (TSA), a specific inhibitor of histone deacetylase (HDAC). The effect of oxamflatin on the transcriptional activity of the cytomegalovirus (CMV) promoter was examined and compared with known HDAC inhibitors, TSA, sodium n-butyrate, and FR901228. Oxamflatin as well as all these inhibitors greatly enhanced the transcriptional activity of the CMV promoter in a dose-dependent manner. Oxamflatin, like TSA, inhibited intracellular HDAC activity, as a result of which marked amounts of acetylated histone species accumulated. Finally, effects on expression of several endogenous genes involved in cell morphology and cell cycle control in HeLa cells were analysed. Expression of gelsolin, cyclin E and Cdk inhibitors including p21WAF1/Cip1 was highly augmented, while that of cyclin A and cyclin D1 was decreased by oxamflatin. These results suggest that changes in the expression pattern of the genes regulating cell morphology and the cell cycle due to histone hyperacetylation are responsible for the antitumor activity, the morphological change and the cell cycle arrest induced by oxamflatin.


Asunto(s)
Antineoplásicos/farmacología , Quinasas CDC2-CDC28 , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Proteínas Proto-Oncogénicas , Animales , División Celular/efectos de los fármacos , Ciclina A/efectos de los fármacos , Ciclina A/genética , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/genética , Ciclinas/efectos de los fármacos , Ciclinas/genética , Ciclinas/metabolismo , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Femenino , Gelsolina/efectos de los fármacos , Gelsolina/genética , Gelsolina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa/citología , Células HeLa/efectos de los fármacos , Humanos , Mamíferos , Melanoma Experimental/tratamiento farmacológico , Ratones , Neoplasias Peritoneales/tratamiento farmacológico , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Regulación hacia Arriba
15.
Mech Dev ; 75(1-2): 67-79, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9739109

RESUMEN

Although betaglycan (TGF-beta type III receptor) is known to enhance TGF-beta ligand binding to its type II receptor in murine lung epithelial cell lines, the biological significance of this phenomenon in the process of lung organogenesis is not understood. Betaglycan gene expression was detected in embryonic murine lungs undergoing branching morphogenesis in ex vivo culture. Antisense betaglycan oligodeoxynucleotides (ODN) resulted in up to 56% stimulation of lung branching morphogenesis in culture, while betaglycan mRNA and protein expression levels were suppressed by 90 and 82%, respectively. Following abrogation of betaglycan expression with antisense oligodeoxynucleotide, embryonic lungs were relatively insensitive to TGF-beta: TGF-beta2 (0.5 ng/ml) and TGF-beta1 (20 ng/ml), respectively, down-regulated lung morphogenesis by 38 and 34% in control cultures, whereas TGF-beta-induced inhibition was attenuated to 13 and 26% respectively, in the presence of betaglycan antisense oligodeoxynucleotides. TGF-beta neutralizing antibodies also prevented TGF-beta-mediated inhibition of lung branching in culture, supporting the speculation that autocrine/paracrine TGF-beta signaling is minimal in the absence of betaglycan. Betaglycan was immunolocalized mainly to the epithelial cells in developing airways, a spatial distribution which overlaps with that of TGF-beta type II receptor. Furthermore, abrogation of endogenous betaglycan gene expression prevented the characteristic down-regulation of cyclin A and surfactant protein C (SP-C) mRNAs by exogenous TGF-beta ligands. These results show that betaglycan expression is essential for optimal TGF-beta signaling during embryonic lung development. We therefore conclude that the abrogation of endogenous betaglycan attenuates endogenous autocrine and/or paracrine TGF-beta-mediated negative regulation of lung organogenesis.


Asunto(s)
Pulmón/embriología , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Anticuerpos/farmacología , Técnicas de Cultivo , Ciclina A/efectos de los fármacos , Ciclina A/genética , Epitelio/química , Femenino , Feto , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inmunohistoquímica , Pulmón/química , Pulmón/efectos de los fármacos , Masculino , Ratones , Morfogénesis/efectos de los fármacos , Pruebas de Neutralización , Oligonucleótidos Antisentido/farmacología , Embarazo , Proteoglicanos/análisis , Proteoglicanos/efectos de los fármacos , Proteolípidos/efectos de los fármacos , Proteolípidos/genética , Surfactantes Pulmonares/efectos de los fármacos , Surfactantes Pulmonares/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Receptores de Factores de Crecimiento Transformadores beta/análisis , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos
16.
FASEB J ; 17(2): 186-93, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12554697

RESUMEN

Neurogenesis, or the production of new neurons, is regulated by physiological and pathological processes including aging, stress, and brain injury. Many mitogenic and trophic factors that regulate proliferation of nonneuronal cells are also involved in neurogenesis. These include vascular endothelial cell growth factor (VEGF), which stimulates the incorporation of bromodeoxyuridine (BrdU) into neuronal precursor cells in vitro and in the adult rat brain in vivo. Using BrdU labeling as an index of cell proliferation, we found that the in vitro neuroproliferative effect of VEGF was associated with up-regulation of E2F family transcription factors, cyclin D1, cyclin E, and cdc25. VEGF also increased nuclear expression of E2F1, E2F2, and E2F3, consistent with regulation of the G1/S phase transition of the cell cycle. The proliferative effect of VEGF was inhibited by the extracellular signal-regulated kinase kinase (MEK) inhibitor PD98059, the phospholipase C (PLC) inhibitor U73122, the protein kinase C (PKC) inhibitor GF102390X, and the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin, indicating involvement of multiple signaling pathways. These findings help to provide a molecular basis for some of the recently identified neuronal effects of VEGF.


Asunto(s)
Proteínas de Ciclo Celular , Corteza Cerebral/efectos de los fármacos , Proteínas de Unión al ADN , Factores de Crecimiento Endotelial/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Linfocinas/farmacología , Factores de Transcripción/efectos de los fármacos , Androstadienos/farmacología , Animales , Western Blotting , Bromodesoxiuridina/metabolismo , División Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Relación Dosis-Respuesta a Droga , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F2 , Factor de Transcripción E2F3 , Electroforesis en Gel de Poliacrilamida , Flavonoides/farmacología , Imidazoles/farmacología , Ratones , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Wortmanina
17.
Clin Cancer Res ; 7(11): 3533-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11705873

RESUMEN

Thymidylate synthase (TS) is responsible for the de novo synthesis of thymidylate, which is required for DNA synthesis and repair and which is an important target for fluoropyrimidines such as 5-fluorouracil (5-FU), and antifolates such as Tomudex (TDX), ZD9331, and multitargeted antifolate (MTA). To study the importance of TS expression in determining resistance to these agents, we have developed an MDA435 breast cancer-derived cell line with tetracycline-regulated expression of TS termed MTS-5. We have demonstrated that inducible expression of TS increased the IC(50) dose of the TS-targeted therapeutic agents 5-FU, TDX, and ZD9331 by 2-, 9- and 24-fold respectively. An IC(50) dose for MTA was unobtainable when TS was overexpressed in these cells, which indicated that MTA toxicity is highly sensitive to increased TS expression levels. The growth inhibitory effects of the chemotherapeutic agents CPT-11, cisplatin, oxaliplatin, and Taxol were unaffected by TS up-regulation. Cell cycle analyses revealed that IC(50) doses of 5-FU, TDX and MTA caused an S-phase arrest in cells that did not overexpress TS, and this arrest was overcome when TS was up-regulated. Furthermore, the S-phase arrest was accompanied by 2- to 4-fold increased expression of the cell cycle regulatory genes cyclin E, cyclin A, and cyclin dependent kinase 2 (cdk2). These results indicate that acute increases in TS expression levels play a key role in determining cellular sensitivity to TS-directed chemotherapeutic drugs by modulating the degree of S-phase arrest caused by these agents. Moreover, CPT-11, cisplatin, oxaliplatin, and Taxol remain highly cytotoxic in cells that overexpress TS.


Asunto(s)
Quinasas CDC2-CDC28 , Camptotecina/análogos & derivados , Tetraciclina/farmacología , Timidilato Sintasa/efectos de los fármacos , Northern Blotting , Western Blotting , Camptotecina/farmacología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Cisplatino/farmacología , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Fluorouracilo/farmacología , Antagonistas del Ácido Fólico/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Irinotecán , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Compuestos Organoplatinos/farmacología , Oxaliplatino , Paclitaxel/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Quinazolinas/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tiofenos/farmacología , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Células Tumorales Cultivadas
18.
Asian J Androl ; 17(6): 996-1005, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26289399

RESUMEN

This study was designed to explore the regulatory effects of male germ cell secreting factor NODAL on Sertoli cell fate decisions from obstructive azoospermia (OA) and nonobstructive azoospermia (NOA) patients. Human Sertoli cells and male germ cells were isolated using two-step enzymatic digestion and SATPUT from testes of azoospermia patients. Expression of NODAL and its multiple receptors in human Sertoli cells and male germ cells were characterized by reverse transcription-polymerase chain reaction (RT-PCR) and immunochemistry. Human recombinant NODAL and its receptor inhibitor SB431542 were employed to probe their effect on the proliferation of Sertoli cells using the CCK-8 assay. Quantitative PCR and Western blots were utilized to assess the expression of Sertoli cell functional genes and proteins. NODAL was found to be expressed in male germ cells but not in Sertoli cells, whereas its receptors ALK4, ALK7, and ACTR-IIB were detected in Sertoli cells and germ cells, suggesting that NODAL plays a regulatory role in Sertoli cells and germ cells via a paracrine and autocrine pathway, respectively. Human recombinant NODAL could promote the proliferation of human Sertoli cells. The expression of cell cycle regulators, including CYCLIN A, CYCLIN D1 and CYCLIN E, was not remarkably affected by NODAL signaling. NODAL enhanced the expression of essential growth factors, including GDNF, SCF, and BMP4, whereas SB431542 decreased their levels. There was not homogeneity of genes changes by NODAL treatment in Sertoli cells from OA and Sertoli cell-only syndrome (SCO) patients. Collectively, this study demonstrates that NODAL produced by human male germ cells regulates proliferation and numerous gene expression of Sertoli cells.


Asunto(s)
Azoospermia/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Nodal/farmacología , Proteínas Recombinantes/farmacología , Células de Sertoli/efectos de los fármacos , Espermatozoides/metabolismo , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II/metabolismo , Adulto , Comunicación Autocrina , Benzamidas/farmacología , Western Blotting , Proteína Morfogenética Ósea 4/efectos de los fármacos , Proteína Morfogenética Ósea 4/metabolismo , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Dioxoles/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Inmunohistoquímica , Masculino , Proteína Nodal/metabolismo , Comunicación Paracrina , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/efectos de los fármacos , Factor de Células Madre/metabolismo
19.
J Invest Dermatol ; 114(3): 581-6, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10692121

RESUMEN

CD40 is expressed in normal human keratinocytes, especially in the basal cell layer. We have recently reported that CD40 ligation strongly inhibits keratinocyte proliferation and induces their differentiation. In this study, the CD40 pathway that prevents keratinocyte growth was investigated. We first reported that interferon-gamma treatment potentiated the CD40-mediated inhibition of keratinocyte proliferation. CD40-CD40 ligand interactions, in the presence or absence of interferon-gamma, neither enhanced spontaneous keratinocyte apoptosis, nor did it enhance apoptosis induced by various agents. More importantly, we showed that CD40 signaling altered the keratinocyte cell cycle, as demonstrated by a decreasing number of cells in the G1 and S phases and an accumulation in G2/M phase of the cell cycle. Furthermore, western blot analysis of cell cycle regulatory proteins, showed a decrease in cyclin A and E expression in CD40-activated keratinocytes. Collectively, these results indicate that CD40 ligation inhibits keratinocyte renewal by a mechanism independent of cell apoptosis and that modulation of the keratinocyte cell cycle is an additional outcome of CD40 signaling.


Asunto(s)
Antígenos CD40/farmacología , Queratinocitos/citología , Glicoproteínas de Membrana/farmacología , Apoptosis/inmunología , Ligando de CD40 , Ciclo Celular/efectos de los fármacos , Ciclo Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Ciclina A/biosíntesis , Ciclina A/efectos de los fármacos , Ciclina E/biosíntesis , Ciclina E/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Interfase/efectos de los fármacos , Ligandos , Fase S
20.
Cancer Lett ; 159(2): 151-8, 2000 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-10996726

RESUMEN

In this study, we examined whether or not a small peptide derived from p16(INK4A) protein with the antennapedia carrier sequence could inhibit the growth of pancreatic cancer cells through the inhibition of cell cycle progression. Growth inhibition by the p16-derived peptide was observed in a time- and dose-dependent manner in AsPC-1 and BxPC-3 cells (p16-negative and pRb-positive), whereas Saos-2 cells (p16-positive and pRb-negative) showed no inhibitory effect. In AsPC-1 and BxPC-3 cells, the proportion of cells in the G(1) phase markedly increased 48 h after treatment with 20 microM p16-derived peptide. Cell-cycle analysis of Saos-2 cells showed little change during the entire period of treatment. Immunoblot analysis showed inhibition of pRb phosphorylation after treatment of BxPC-3 with 10 microM p16 peptide. Furthermore, the p16 peptide caused a decrease in cyclin A at later times of treatment. These results demonstrate that the p16-derived peptide can inhibit the growth of p16-negative and pRb-positive pancreatic cancer cells by means of G(1) phase cell cycle arrest resulting from the inhibition of pRb phosphorylation. Restoration of p16/pRb tumor-suppressive pathway by re-expression of p16(INK4A) may play a therapeutic role in the treatment of pancreatic cancer.


Asunto(s)
Proteínas Portadoras/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Homeodominio/farmacología , Proteínas Nucleares , Neoplasias Pancreáticas/prevención & control , Proteína de Retinoblastoma/efectos de los fármacos , Factores de Transcripción , Secuencia de Aminoácidos , Proteína con Homeodominio Antennapedia , Biotinilación , Proteínas Portadoras/síntesis química , División Celular/efectos de los fármacos , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Relación Dosis-Respuesta a Droga , Proteínas de Homeodominio/síntesis química , Humanos , Datos de Secuencia Molecular , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA