Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Cancer Cell ; 11(3): 275-89, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17349584

RESUMEN

The FEZ1/LZTS1 (LZTS1) protein is frequently downregulated in human cancers of different histotypes. LZTS1 is expressed in normal tissues, and its introduction in cancer cells inhibits cell growth and suppresses tumorigenicity, owing to an accumulation of cells in G2/M. Here, we define its role in cell cycle regulation and tumor progression by generating Lzts1 knockout mice. In Lzts1(-/-) mouse embryo fibroblasts (MEFs), Cdc25C degradation was increased during M phase, resulting in decreased Cdk1 activity. As a consequence, Lzts1(-/-) MEFs showed accelerated mitotic progression, resistance to taxol- and nocodazole-induced M phase arrest, and improper chromosome segregation. Accordingly, Lzts1 deficiency was associated with an increased incidence of both spontaneous and carcinogen-induced cancers in mice.


Asunto(s)
Proteína Quinasa CDC2/fisiología , Proteínas de Ciclo Celular/fisiología , Transformación Celular Neoplásica , Mitosis , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/fisiología , Fosfatasas cdc25/fisiología , Animales , Antineoplásicos/farmacología , Carcinógenos , División Celular , Células Cultivadas , Segregación Cromosómica , Dimetilnitrosamina/análogos & derivados , Fibroblastos/metabolismo , Fibroblastos/patología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Nocodazol/farmacología , Paclitaxel/farmacología , Neoplasias Gástricas/inducido químicamente , Neoplasias Gástricas/genética , Proteínas Supresoras de Tumor/genética
2.
Curr Opin Cell Biol ; 18(2): 185-91, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16488126

RESUMEN

The CDC25 phosphatases are key regulators of normal cell division and the cell's response to DNA damage. Earlier studies suggested non-overlapping roles for each isoform during a specific cell cycle phase. However, recent data suggest that multiple CDC25 isoforms cooperate to regulate each cell cycle transition. For instance, although CDC25A was initially thought to exclusively regulate the G(1)-S transition, recent data demonstrate a significant role for CDC25A in the G(2)-M transition. Further evidence demonstrates that in addition to the ATM/ATR-CHK pathway, a p38-MAPKAP pathway is also involved in controlling CDC25 activity during G(2)/M checkpoint activation. Together with the fact that CDC25 overexpression is reported in many cancers, these data highlight the significance of developing specific CDC25 inhibitors for cancer therapy.


Asunto(s)
Ciclo Celular/fisiología , Fosfatasas cdc25/fisiología , Animales , Benzoquinonas/uso terapéutico , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Isoenzimas/fisiología , Modelos Biológicos , Naftoquinonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Quinolonas/uso terapéutico , Quinonas/uso terapéutico , Tiazoles/uso terapéutico , Fosfatasas cdc25/antagonistas & inhibidores , Fosfatasas cdc25/metabolismo
3.
Nat Genet ; 30(4): 446-9, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11912493

RESUMEN

In a wide variety of animal species, oocyte maturation is arrested temporarily at prophase of meiosis I (ref. 1). Resumption of meiosis requires activation of cyclin-dependent kinase-1 (CDK1, p34cdc2), one component of maturation-promoting factor (MPF). The dual specificity phosphatases Cdc25a, Cdc25b and Cdc25c are activators of cyclin-dependent kinases; consequently, they are postulated to regulate cell-cycle progression in meiosis and mitosis as well as the DNA-damage response. We generated Cdc25b-deficient (Cdc25b-/-) mice and found that they are viable. As compared with wildtype cells, fibroblasts from Cdc25b-/- mice grew vigorously in culture and arrested normally in response to DNA damage. Female Cdc25b-/- mice were sterile, and Cdc25b-/- oocytes remained arrested at prophase with low MPF activity. Microinjection of wildtype Cdc25b mRNA into Cdc25b-/- oocytes caused activation of MPF and resumption of meiosis. Thus, Cdc25b-/- female mice are sterile because of permanent meiotic arrest resulting from the inability to activate MPF. Cdc25b is therefore essential for meiotic resumption in female mice. Mice lacking Cdc25b provide the first genetic model for studying the mechanisms regulating prophase arrest in vertebrates.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Meiosis , Oocitos/fisiología , Fosfatasas cdc25/genética , Fosfatasas cdc25/fisiología , Animales , Southern Blotting , Western Blotting , Células Cultivadas , Daño del ADN , Femenino , Sistema de Señalización de MAP Quinasas , Masculino , Mesotelina , Ratones , Microscopía Fluorescente , Mitosis , Modelos Genéticos , Oocitos/metabolismo , ARN Mensajero/metabolismo
4.
Carcinogenesis ; 33(9): 1754-61, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22764135

RESUMEN

Cell division cycle 25A (CDC25A) is a dual-specificity phosphatase that removes inhibitory phosphates from cyclin-dependent kinases, allowing cell-cycle progression. Activation of cell-cycle checkpoints following DNA damage results in the degradation of CDC25A, leading to cell-cycle arrest. Ultraviolet (UV) irradiation, which causes most skin cancer, results in both DNA damage and CDC25A degradation. We hypothesized that ablation of CDC25A in the skin would increase cell-cycle arrest following UV irradiation, allowing for improved repair of DNA damage and decreased tumorigenesis. Cdc25a(fl/fl) /Krt14-Cre recombinase mice, with decreased CDC25A in the epithelium of the skin, were generated and exposed to UV. UV-induced DNA damage, in the form of cyclopyrimidine dimers and 8-oxo-deoxyguanosine adducts, was eliminated earlier from CDC25A-deficient epidermis. Surprisingly, loss of CDC25A did not alter epidermal proliferation or cell cycle after UV exposure. However, the UV-induced apoptotic response was prolonged in CDC25A-deficient skin. Double labeling of cleaved caspase-3 and the DNA damage marker γH2A.X revealed many of the apoptotic cells in UV-exposed Cdc25a mutant skin had high levels of DNA damage. Induction of skin tumors by UV irradiation of Cdc25a mutant and control mice on a skin tumor susceptible to v-ras(Ha) Tg.AC mouse background revealed UV-induced papillomas in Cdc25a mutants were significantly smaller than in controls in the first 6 weeks following UV exposure, although there was no difference in tumor multiplicity or incidence. Thus, deletion of Cdc25a increased apoptosis and accelerated the elimination of DNA damage following UV but did not substantially alter cell-cycle regulation or tumorigenesis.


Asunto(s)
Apoptosis/efectos de la radiación , Daño del ADN , ADN/efectos de la radiación , Piel/efectos de la radiación , Fosfatasas cdc25/fisiología , Animales , Ciclo Celular , Proliferación Celular/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Piel/patología , Rayos Ultravioleta , Fosfatasas cdc25/deficiencia
5.
Carcinogenesis ; 33(12): 2344-50, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22962304

RESUMEN

Human SERPINB5, commonly known as maspin, has diverse functions as a tumor suppressor. In this study, we discovered that maspin has a novel role in cell cycle control, and common variants were discovered to be associated with gastric cancer. The genotypes of 836 unrelated Korean participants (including 430 with gastric cancer) were examined for 12 tag single-nucleotide polymorphisms (SNPs) and imputed for 178 SNPs in the maspin gene. Susceptibility to diffuse-type gastric cancer was strongly and significantly associated with several SNPs including rs3744941 (C>T) in the promoter (TT versus CC+CT, odds ratio = 0.56 [0.37-0.83], P = 0.0038) and rs8089104 (C>T) in intron 1 (TT+CT versus CC, odds ratio = 1.7 [1.2-2.5], P = 0.0021). No SNPs were associated with susceptibility to intestinal-type gastric cancer. A haplotype of three highly correlated promoter SNPs associated with higher cancer risk showed 40% of the activity of a non-risk-associated haplotype promoter in the diffuse-type gastric cancer cell line MKN45. Maspin downregulation achieved either by a short hairpin RNA targeting maspin or overexpression of the E2F1-DP1 complex in MKN45 cells dramatically accelerated cell cycle progression and caused an increase of active CDC25C levels and a decrease of inactive CDK1 levels. In contrast, maspin upregulation had the opposite effect, substantially retarding cell proliferation. Therefore, our results suggest that a maspin promoter haplotype that reduces maspin gene expression accelerates cell cycle progression and, consequently, is associated with increased susceptibility to diffuse-type gastric cancer. Furthermore, a novel maspin-related pathway is demonstrated to underlie gastric carcinogenesis.


Asunto(s)
Ciclo Celular , Serpinas/fisiología , Neoplasias Gástricas/patología , Adulto , Anciano , Proteína Quinasa CDC2/fisiología , Factor de Transcripción E2F1/fisiología , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Neoplasias Gástricas/genética , Fosfatasas cdc25/fisiología
6.
Cancer Cell ; 3(3): 247-58, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12676583

RESUMEN

Chk1 kinase coordinates cell cycle progression and preserves genome integrity. Here, we show that chemical or genetic ablation of human Chk1 triggered supraphysiological accumulation of the S phase-promoting Cdc25A phosphatase, prevented ionizing radiation (IR)-induced degradation of Cdc25A, and caused radioresistant DNA synthesis (RDS). The basal turnover of Cdc25A operating in unperturbed S phase required Chk1-dependent phosphorylation of serines 123, 178, 278, and 292. IR-induced acceleration of Cdc25A proteolysis correlated with increased phosphate incorporation into these residues generated by a combined action of Chk1 and Chk2 kinases. Finally, phosphorylation of Chk1 by ATM was required to fully accelerate the IR-induced degradation of Cdc25A. Our results provide evidence that the mammalian S phase checkpoint functions via amplification of physiologically operating, Chk1-dependent mechanisms.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Quinasas/metabolismo , Fosfatasas cdc25/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Replicación del ADN/efectos de la radiación , Proteínas de Unión al ADN , Activación Enzimática , Células HeLa , Humanos , Cinética , Modelos Biológicos , Fosforilación , Proteínas Serina-Treonina Quinasas/fisiología , Radiación Ionizante , Fase S/efectos de la radiación , Serina/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Fosfatasas cdc25/efectos de la radiación
7.
Curr Opin Cell Biol ; 13(6): 738-47, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11698191

RESUMEN

The ability to preserve genomic integrity is a fundamental feature of life. Recent findings regarding the molecular basis of the cell-cycle checkpoint responses of mammalian cells to genotoxic stress have converged into a two-wave concept of the G1 checkpoint, and shed light on the so-far elusive intra-S-phase checkpoint. Rapidly operating cascades that target the Cdc25A phosphatase appear central in both the initiation wave of the G1 checkpoint (preceding the p53-mediated maintenance wave) and the transient intra-S-phase response. Multiple links between defects in the G1/S checkpoints, genomic instability and oncogenesis are emerging, as are new challenges and hopes raised by this knowledge.


Asunto(s)
Daño del ADN , Fase G1 , Fase S , Animales , Predicción , Modelos Biológicos , Neoplasias/genética , Proteínas Quinasas/fisiología , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/fisiología , Fosfatasas cdc25/fisiología
8.
Trends Cell Biol ; 16(6): 285-92, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16682204

RESUMEN

Members of the eukaryotic Cdc25 phosphatase family are key targets of the Chk1 and Chk2 checkpoint kinases, which inactivate Cdc25 to halt cell cycle progression when DNA is damaged or incompletely replicated. Now, new kinases that phosphorylate and inactivate Cdc25 are being discovered, including MAPKAP kinase-2, a component of the p38 stress-activated MAP kinase pathway. The roles of other kinases, such as cyclin-dependent kinase, Polo and Aurora A kinase, in controlling the localization or the activation of Cdc25, are controversial. Here, we discuss new data that suggests that different Cdc25 isoforms and regulators of Cdc25 are differentially required for normal cell cycle progression and recovery from checkpoint arrest.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Fosfatasas cdc25/fisiología , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular/fisiología , Quinasa de Punto de Control 2 , Daño del ADN , Humanos , Péptidos y Proteínas de Señalización Intracelular , Modelos Genéticos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/fisiología , Huso Acromático/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Nat Cell Biol ; 6(9): 884-91, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15311285

RESUMEN

Entry into mitosis occurs after activation of Cdk1, resulting in chromosome condensation in the nucleus and centrosome separation, as well as increased microtubule nucleation activity in the cytoplasm. The active cyclin-B1-Cdk1 complex first appears at the centrosome, suggesting that the centrosome may facilitate the activation of mitotic regulators required for the commitment of cells to mitosis. However, the signalling pathways involved in controlling the initial activation of Cdk1 at the centrosome remain largely unknown. Here, we show that human Chk1 kinase localizes to interphase, but not mitotic, centrosomes. Chemical inhibition of Chk1 resulted in premature centrosome separation and activation of centrosome-associated Cdk1. Forced immobilization of kinase-inactive Chk1 to centrosomes also resulted in premature Cdk1 activation. Conversely, under such conditions wild-type Chk1 impaired activation of centrosome-associated Cdk1, thereby resulting in DNA endoreplication and centrosome amplification. Activation of centrosomal Cdk1 in late prophase seemed to be mediated by cytoplasmic Cdc25B, whose activity is controlled by centrosome-associated Chk1. These results suggest that centrosome-associated Chk1 shields centrosomal Cdk1 from unscheduled activation by cytoplasmic Cdc25B, thereby contributing to proper timing of the initial steps of cell division, including mitotic spindle formation.


Asunto(s)
Centrosoma/enzimología , Ciclinas/metabolismo , Proteínas Quinasas/fisiología , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/fisiología , Proteínas de Ciclo Celular/fisiología , División Celular , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina B/metabolismo , Ciclina B1 , Activación Enzimática , Humanos , Interfase , Microscopía Confocal , Unión Proteica , Proteínas Quinasas/metabolismo , Huso Acromático , Fosfatasas cdc25/fisiología
10.
Dev Biol ; 326(1): 131-42, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19063878

RESUMEN

Following fertilization of many animal embryos, rapid synchronous cleavage divisions give way to longer, asynchronous cell cycles at the midblastula transition (MBT). The cell cycle changes at the MBT, including the addition of gap phases and checkpoint controls, are accompanied by activation of the zygotic genome and the onset of cell motility. Whereas the biochemical changes accompanying the MBT in the vertebrate embryo have been extensively documented, the cellular events are not well understood. We show that cell cycle remodeling during the zebrafish MBT includes the transcription-independent acquisition of a G2 phase that is essential for preventing entry into mitosis before S-phase completion in cycles 11-13. We provide evidence from high-resolution imaging that inhibition of Cdc25a and Cdk1 activity, but not Cdk2 activity, is essential for cell cycle lengthening and asynchrony between cycles 9 and 12. We demonstrate that lengthening is not required for initiation of zygotic transcription. Our results are consistent with findings from Drosophila and Xenopus that indicate the central importance of G2 addition in checkpoint establishment, and point to similar mechanisms governing the MBT in diverse species.


Asunto(s)
Blástula/fisiología , Fase G2/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Proteína Quinasa CDC2/fisiología , Ciclo Celular/fisiología , Quinasa 2 Dependiente de la Ciclina/fisiología , Activación Transcripcional/fisiología , Fosfatasas cdc25/fisiología
11.
J Cell Biol ; 171(1): 35-45, 2005 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16216921

RESUMEN

Cdc25 phosphatases are essential for the activation of mitotic cyclin-Cdks, but the precise roles of the three mammalian isoforms (A, B, and C) are unclear. Using RNA interference to reduce the expression of each Cdc25 isoform in HeLa and HEK293 cells, we observed that Cdc25A and -B are both needed for mitotic entry, whereas Cdc25C alone cannot induce mitosis. We found that the G2 delay caused by small interfering RNA to Cdc25A or -B was accompanied by reduced activities of both cyclin B1-Cdk1 and cyclin A-Cdk2 complexes and a delayed accumulation of cyclin B1 protein. Further, three-dimensional time-lapse microscopy and quantification of Cdk1 phosphorylation versus cyclin B1 levels in individual cells revealed that Cdc25A and -B exert specific functions in the initiation of mitosis: Cdc25A may play a role in chromatin condensation, whereas Cdc25B specifically activates cyclin B1-Cdk1 on centrosomes.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/fisiología , Centrosoma/química , Quinasas Ciclina-Dependientes/metabolismo , Mitosis/efectos de los fármacos , Fosfatasas cdc25/metabolismo , Fosfatasas cdc25/fisiología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/farmacología , Línea Celular , División del Núcleo Celular/efectos de los fármacos , Centrosoma/efectos de los fármacos , Centrosoma/metabolismo , Ciclina B1 , Quinasas Ciclina-Dependientes/análisis , Quinasas Ciclina-Dependientes/efectos de los fármacos , Activación Enzimática , Células HeLa , Humanos , Mitosis/fisiología , ARN Interferente Pequeño/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Fosfatasas cdc25/farmacología
12.
Biol Pharm Bull ; 33(7): 1112-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606298

RESUMEN

We have recently demonstrated that TRB3, a novel stress-inducible protein, is an unstable protein regulated by the ubiquitin-proteasome system. The expression level of TRB3 protein is down-regulated by anaphase-promoting complex/cyclosome-cell division cycle division 20 homolog 1 (APC/C(Cdh1)) through its D-box motif. Here we demonstrate that TRB3 regulates the stability of cell division cycle 25 A (Cdc25A), an essential activator of cyclin dependent kinases (CDKs). The expression level of Cdc25A protein is suppressed by over-expression of TRB3, while knockdown of TRB3 enhances the endogenous Cdc25A expression level. On the other hand, Cdc25A degradation induced by DNA damage is significantly rescued by TRB3. When serine residues in the DSG motif, which is the critical sequences for the degradation of Cdc25A induced by DNA damage, is mutated to alanine (Cdc25A(DSG2X)), both stimulatory and protective effects of TRB3 on the Cdc25A degradation is disappeared. TRB3 protein interacts with both wild Cdc25A and mutant Cdc25A(DSG2X). Expression level of the endogenous TRB3 protein is down-regulated in a genotoxic condition. These results suggest TRB3 is a regulator for adjusting the expression level of Cdc25A both in a normal and a genotoxic conditions.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Represoras/fisiología , Fosfatasas cdc25/fisiología , Western Blotting , Línea Celular , Daño del ADN , Humanos , Inmunoprecipitación , Interferencia de ARN
13.
J Virol ; 82(9): 4527-32, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18272575

RESUMEN

Earlier studies have shown that in herpes simplex virus 1-infected cells, ICP22 upregulates the accumulation of a subset of gamma(2) proteins exemplified by the products of the U(L)38, U(L)41, and U(S)11 genes. The ICP22-dependent process involves degradation of cyclins A and B1, the stabilization and activation of cdc2, physical interaction of activated cdc2 with the U(L)42 DNA synthesis processivity factor, and recruitment and phosphorylation of topoisomerase IIalpha by the cdc2/U(L)42 complex. Activation of cdc2, the first step in the process, is a key function of the mitotic phosphatase cdc25C. To define the role of cdc25C, we probed some features of the ICP22-dependent pathway of upregulation of gamma(2) genes in cdc25C(-/-) cells and in cdc25C(+/+) cells derived from sibling mice. We report that cyclin B1 turned over in cdc25C(+/+) or cdc25C(-/-) cells at the same rate, that cdc2 increased in amount, and that U(S)11 and U(L)38 proteins and infectious virus accumulated in smaller amounts than in wild-type infected cells. The reduction in U(L)38 protein accumulation and virus was greater in cdc25C(-/-) cells infected with virus lacking ICP22 than in cells infected with wild-type virus. We conclude that cdc25C phosphatase plays a role in viral replication and that this role extends beyond its function of activating cdc2 for initiation of the ICP22-dependent cascade for upregulation of gamma(2) gene expression.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Replicación Viral/genética , Fosfatasas cdc25/fisiología , Animales , Proteínas de la Cápside , Línea Celular , Células Cultivadas , Humanos , Ratones , Proteínas de Unión al ARN , Regulación hacia Arriba , Proteínas Virales/genética
14.
J Cell Biol ; 147(7): 1371-8, 1999 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-10613895

RESUMEN

Centrosome duplication is marked by discrete changes in centriole structure that occur in lockstep with cell cycle transitions. We show that mitotic regulators govern steps in centriole replication in Drosophila embryos. Cdc25(string), the expression of which initiates mitosis, is required for completion of daughter centriole assembly. Cdc20(fizzy), which is required for the metaphase-anaphase transition, is required for timely disengagement of mother and daughter centrioles. Stabilization of mitotic cyclins, which prevents exit from mitosis, blocks assembly of new daughter centrioles. Common regulation of the nuclear and centrosome cycles by mitotic regulators may ensure precise duplication of the centrosome.


Asunto(s)
Centrosoma/fisiología , Proteínas de Drosophila , Mitosis/fisiología , Proteínas Tirosina Fosfatasas , Proteínas de Saccharomyces cerevisiae , Secuencia de Aminoácidos , Animales , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Centriolos/genética , Centriolos/fisiología , Ciclinas/fisiología , Drosophila , Datos de Secuencia Molecular , Fosfoproteínas Fosfatasas/fisiología , Fosfatasas cdc25/fisiología
15.
Int J Radiat Biol ; 85(2): 167-74, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19280470

RESUMEN

PURPOSE: To determine the efficacy of a caffeine derivative 1-methylxanthine (1-MTX) in increasing radiosensitivity of cancer cells and elucidate the underlying mechanisms in vitro. MATERIALS AND METHODS: RKO human colorectal cancer cells carrying wild type protein 53 kDa (p53) were incubated with 3 mM 1-MTX for 30 min, exposed to 4 Gy ionizing radiation, and further incubated with 1-MTX for three days. The clonogenic cell death was determined, and the cell cycle distribution and apoptosis were studied with flow cytometry at different times after irradiation. The DNA double strand break (DNA DSB) was examined using phosphorylated Histone2A (gamma-H2AX) foci formation, and the expression/activity of checkpoint 2 kinase (Chk2), cell division cycle 25 (Cdc25) phosphatase and cyclin B1/Cdc2 kinase were also investigated using western blotting and in vitro kinase assays. RESULTS: The treatment with 3 mM 1-MTX increased the radiation-induced clonogenic and apoptotic cell death. The radiation-induced phosphorylation of Chk2 and Cdc25c and the radiation-induced increase in the cyclin B1/Cdc2 kinas activity were little affected by 1-MTX. The radiation-induced G2/M arrest was only slightly shortened and the expression of radiation-induced gamma-H2AX was markedly prolonged by 1-MTX. CONCLUSIONS: 1-MTX significantly increased the radiosensitivity of RKO human colorectal cancer cells carrying wild type p53 mainly by inhibiting the repair of radiation-induced DNA DSB without causing significant alteration in radiation-induced G2/M arrest. Such a radiosensitization occurred at 1-MTX concentrations almost non-toxic to the target tumor cells.


Asunto(s)
Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Xantinas/farmacología , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Daño del ADN , Fase G2/efectos de la radiación , Histonas/análisis , Humanos , Proteína p53 Supresora de Tumor/análisis , Fosfatasas cdc25/fisiología
16.
Cancer Res ; 67(14): 6605-11, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638870

RESUMEN

CDC25A phosphatase activates multiple cyclin-dependent kinases (CDK) during cell cycle progression. Inactivation of CDC25A by ubiquitin-mediated degradation is a major mechanism of DNA damage-induced S-G(2) checkpoint. Although increased CDC25A expression has been reported in various human cancer tissues, it remains unclear whether CDC25A activation is a critical rate-limiting step of carcinogenesis. To assess the role for CDC25A in cell cycle control and carcinogenesis, we used a Cdc25A-null mouse strain we recently generated. Whereas Cdc25A(-/-) mice exhibit early embryonic lethality, Cdc25A(+/-) mice show no appreciable developmental defect. Cdc25A(+/-) mouse embryonic fibroblasts (MEF) exhibit normal kinetics of cell cycle progression at early passages, modestly enhanced G(2) checkpoint response to DNA damage, and shortened proliferative life span, compared with wild-type MEFs. Importantly, Cdc25A(+/-) MEFs are significantly resistant to malignant transformation induced by coexpression of H-ras(V12) and a dominant negative p53 mutant. The rate-limiting role for CDC25A in transformation is further supported by decreased transformation efficiency in MCF-10A human mammary epithelial cells stably expressing CDC25A small interfering RNA. Consistently, Cdc25A(+/-) mice show substantially prolonged latency in mammary tumorigenesis induced by MMTV-H-ras or MMTV-neu transgene, whereas MMTV-myc-induced tumorigenesis is not significantly affected by Cdc25A heterozygosity. Mammary tissues of Cdc25A(+/-);MMTV-neu mice before tumor development display less proliferative response to the oncogene with increased tyrosine phosphorylation of CDK1/2, but show no significant change in apoptosis. These results suggest that Cdc25A plays a rate-limiting role in transformation and tumor initiation mediated by ras activation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Fosfatasas cdc25/genética , Fosfatasas cdc25/fisiología , Proteínas ras/metabolismo , Animales , Ciclo Celular , Transformación Celular Neoplásica , Células Cultivadas , Fibroblastos/metabolismo , Fase G2 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fase S , Factores de Tiempo
17.
Mol Cancer Ther ; 7(12): 3789-99, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19074854

RESUMEN

Cdc25C is a dual-specificity phosphatase that is involved in induction of mitosis by removal of the inhibitory phosphates from cyclin-dependent kinase 1/cyclin B. In this study, adenovirus-mediated overexpression of Cdc25C sensitizes U2OS tumor cells to doxorubicin-induced apoptosis. U2OS cells that stably overexpress Cdc25C are also sensitized to doxorubicin-induced cell death. These cells show reduced phosphorylation of cyclin-dependent kinase 1 on Tyr15 and impaired up-regulation of p21 in response to treatment with doxorubicin. In contrast to doxorubicin, overexpression of Cdc25C does not confer sensitivity to apoptosis on treatment with 5-fluorouracil or hydroxyurea. This sensitization of tumor cells to doxorubicin-induced cell death by overexpression of Cdc25C is not p53 dependent. Intriguingly, nontransformed MCF10A cells are not sensitized to doxorubicin treatment by overexpression of Cdc25C nor does the lack of Cdc25C affect cell cycle progression or the G2 arrest caused by doxorubicin. These results support the idea that a combination of overexpressing Cdc25C with treatment with conventional genotoxic agents should be given serious considerations as a novel therapeutic strategy.


Asunto(s)
Apoptosis , Doxorrubicina/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Fosfatasas cdc25/fisiología , Antibióticos Antineoplásicos/farmacología , Ciclo Celular , Muerte Celular , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/metabolismo , Fluorouracilo/farmacología , Humanos , Hidroxiurea/farmacología , Fosforilación , ARN Interferente Pequeño/metabolismo , Fosfatasas cdc25/metabolismo
18.
Neural Dev ; 14(1): 7, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30867016

RESUMEN

In the developing neural tube in chicken and mammals, neural stem cells proliferate and differentiate according to a stereotyped spatiotemporal pattern. Several actors have been identified in the control of this process, from tissue-scale morphogens patterning to intrinsic determinants in neural progenitor cells. In a previous study (Bonnet et al. eLife 7, 2018), we have shown that the CDC25B phosphatase promotes the transition from proliferation to differentiation by stimulating neurogenic divisions, suggesting that it acts as a maturating factor for neural progenitors. In this previous study, we set up a mathematical model linking fixed progenitor modes of division to the dynamics of progenitors and differentiated populations. Here, we extend this model over time to propose a complete dynamical picture of this process. We start from the standard paradigm that progenitors are homogeneous and can perform any type of divisions (proliferative division yielding two progenitors, asymmetric neurogenic divisions yielding one progenitor and one neuron, and terminal symmetric divisions yielding two neurons). We calibrate this model using data published by Saade et al. (Cell Reports 4, 2013) about mode of divisions and population dynamics of progenitors/neurons at different developmental stages. Next, we explore the scenarios in which the progenitor population is actually split into two different pools, one of which is composed of cells that have lost the capacity to perform proliferative divisions. The scenario in which asymmetric neurogenic division would induce such a loss of proliferative capacity appears very relevant.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Modelos Biológicos , Células-Madre Neurales/fisiología , Tubo Neural/citología , Tubo Neural/crecimiento & desarrollo , Médula Espinal/citología , Médula Espinal/crecimiento & desarrollo , Fosfatasas cdc25/fisiología , Animales
19.
Nutr Cancer ; 60 Suppl 1: 51-60, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19003581

RESUMEN

Withaferin A (WA) is derived from the medicinal plant Withania somnifera that has been safely used for centuries in the Indian Ayurvedic medicine for treatment of various ailments. We now demonstrate that WA treatment causes G2 and mitotic arrest in human breast cancer cells. Treatment of MDA-MB-231 (estrogen-independent) and MCF-7 (estrogen-responsive) cell lines with WA resulted in a concentration- and time-dependent increase in G2-M fraction, which correlated with a decrease in levels of cyclin-dependent kinase 1 (Cdk1), cell division cycle 25C (Cdc25C) and/or Cdc25B proteins, leading to accumulation of Tyrosine15 phosphorylated (inactive) Cdk1. Ectopic expression of Cdc25C conferred partial yet significant protection against WA-mediated G2-M phase cell cycle arrest in MDA-MB-231 cells. The WA-treated MDA-MB-231 and MCF-7 cells were also arrested in mitosis as judged by fluorescence microscopy and analysis of Ser10 phosphorylated histone H3. Mitotic arrest resulting from exposure to WA was accompanied by an increase in the protein level of anaphase promoting complex/cyclosome substrate securin. In conclusion, the results of this study suggest that G2-M phase cell cycle arrest may be an important mechanism in antiproliferative effect of WA against human breast cancer cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , División Celular/efectos de los fármacos , Ergosterol/análogos & derivados , Fase G2/efectos de los fármacos , Medicina Ayurvédica , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ergosterol/farmacología , Femenino , Histonas/metabolismo , Humanos , Mitosis/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Witanólidos , Fosfatasas cdc25/análisis , Fosfatasas cdc25/fisiología
20.
Trends Plant Sci ; 11(10): 474-9, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16949857

RESUMEN

Progression through the cell cycle is regulated by cyclin-dependent kinases (CDKs). Plants possess a unique class of CDKs, designated B-type CDKs, but seem to lack a functional CDC25 phosphatase, which is a crucial activator of the onset of mitosis in non-plant species. Based on a striking number of functional parallels between the Arabidopsis thaliana CDKB1;1 and the Drosophila melanogaster CDC25 (string), we hypothesize that the acquisition of B-type CDKs and the disappearance of CDC25 in plants might have been associated; in these coupled events, the CDC25-controlled onset of mitosis might have been evolutionarily replaced by a B-type CDK-dominated pathway, eventually resulting in the loss of the CDC25 gene.


Asunto(s)
Quinasas Ciclina-Dependientes/fisiología , Evolución Molecular , Plantas/enzimología , Fosfatasas cdc25/fisiología , Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/clasificación , Quinasas Ciclina-Dependientes/genética , Regulación de la Expresión Génica de las Plantas , Fosforilación , Células Vegetales , Desarrollo de la Planta , Fosfatasas cdc25/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda