Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Cycle ; 18(21): 2986-2997, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31530151

RESUMEN

Calcium (Ca2+)/calmodulin-dependent protein kinase II (CaMKII) had been reported to play a role in the process of fertilization. However, the role of CaMKII in the release of diplotene-arrested oocytes is poorly understood. In this study, we explored the potential effect of CaMKII on Akt1 and the relationship among CaMKII, Akt1 and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) during the meiotic resumption of mouse oocytes. We found that inhibition of CaMKII aggravated diplotene arrest. We detected the expression and distribution of pCaMKII (Thr286), pAkt1 (Ser473), Cdc25B and pCdc2 (Tyr15) when oocytes were treated with KN-93, SH-6, LY294002 or PIP3, respectively. Our data showed that down-regulated CaMKII by KN-93 decreased the levels of pAkt1 (Ser473) and rearranged the distribution of pAkt1 (Ser473). Meanwhile, down-regulated pAkt1 (Ser473) by SH-6 also decreased the levels of pCaMKII (Thr286), Cdc25B and pCdc2 (Tyr15) significantly and rearranged the distributions of pCaMKII (Thr286). Furthermore, our data showed that exogenous PIP3 up-regulated GVBD rates significantly and increased the levels of pCaMKII (Thr286) and pAkt1 (Ser473). On the contrary, down-regulation of PIP3 by LY294002 decreased GVBD rates and the levels of pCaMKII (Thr286) and pAkt1 (Ser473), respectively. Our results showed that Akt1 and CaMKII regulated each other, and PIP3 may be involved in these regulations during the release of mouse oocytes from diplotene arrest.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Profase Meiótica I/fisiología , Oocitos/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proteína Quinasa CDC2/biosíntesis , Femenino , Ratones , Oocitos/crecimiento & desarrollo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Fosfatasas cdc25/biosíntesis
2.
Toxins (Basel) ; 12(1)2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31906179

RESUMEN

Ochratoxin A (OTA), an important mycotoxin that occurs in food and animal feed, has aroused widespread concern in recent years. Previous studies have indicated that OTA causes nephrotoxicity, hepatotoxicity, genotoxicity, immunotoxicity, cytotoxicity, and neurotoxicity. The intestinal toxicity of OTA has gradually become a focus of research, but the mechanisms underlying this toxicity have not been described. Here, differentiated Caco-2 cells were incubated for 48 h with different concentrations of OTA and transcriptome analysis was used to estimate damage to the intestinal barrier. Gene expression profiling was used to compare the characteristics of differentially expressed genes (DEGs). There were altogether 10,090 DEGs, mainly clustered into two downregulation patterns. The Search Tool for Retrieval of Interacting Genes (STRING), which was used to analyze the protein-protein interaction network, indicated that 24 key enzymes were mostly responsible for regulating cell apoptosis. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis was used to validate eight genes, three of which were key genes (CASP3, CDC25B, and EGR1). The results indicated that OTA dose-dependently induces apoptosis in differentiated Caco-2 cells. Transcriptome analysis showed that the impairment of intestinal function caused by OTA might be partly attributed to apoptosis, which is probably associated with downregulation of murine double minute 2 (MDM2) expression and upregulation of Noxa and caspase 3 (CASP3) expression. This study has highlighted the intestinal toxicity of OTA and provided a genome-wide view of biological responses, which provides a theoretical basis for enterotoxicity and should be useful in establishing a maximum residue limit for OTA.


Asunto(s)
Apoptosis/efectos de los fármacos , Ocratoxinas/farmacología , Transcriptoma/efectos de los fármacos , Células CACO-2 , Caspasa 3/biosíntesis , Caspasa 3/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Perfilación de la Expresión Génica , Humanos , Intestinos/patología , Familia de Multigenes , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética
3.
PLoS Genet ; 13(2): e1006564, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28222105

RESUMEN

Cell proliferation has generally been considered dispensable for anteroposterior extension of embryonic axis during vertebrate gastrulation. Signal transducer and activator of transcription 3 (Stat3), a conserved controller of cell proliferation, survival and regeneration, is associated with human scoliosis, cancer and Hyper IgE Syndrome. Zebrafish Stat3 was proposed to govern convergence and extension gastrulation movements in part by promoting Wnt/Planar Cell Polarity (PCP) signaling, a conserved regulator of mediolaterally polarized cell behaviors. Here, using zebrafish stat3 null mutants and pharmacological tools, we demonstrate that cell proliferation contributes to anteroposterior embryonic axis extension. Zebrafish embryos lacking maternal and zygotic Stat3 expression exhibit normal convergence movements and planar cell polarity signaling, but transient axis elongation defect due to insufficient number of cells resulting largely from reduced cell proliferation and increased apoptosis. Pharmacologic inhibition of cell proliferation during gastrulation phenocopied axis elongation defects. Stat3 regulates cell proliferation and axis extension in part via upregulation of Cdc25a expression during oogenesis. Accordingly, restoring Cdc25a expression in stat3 mutants partially suppressed cell proliferation and gastrulation defects. During later development, stat3 mutant zebrafish exhibit stunted growth, scoliosis, excessive inflammation, and fail to thrive, affording a genetic tool to study Stat3 function in vertebrate development, regeneration, and disease.


Asunto(s)
Proliferación Celular/genética , Desarrollo Embrionario/genética , Factor de Transcripción STAT3/genética , Proteínas de Pez Cebra/genética , Fosfatasas cdc25/genética , Animales , Polaridad Celular/genética , Gástrula/crecimiento & desarrollo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Morfogénesis/genética , Proteínas Mutantes/genética , Factor de Transcripción STAT3/biosíntesis , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/biosíntesis , Fosfatasas cdc25/biosíntesis
4.
Int J Mol Sci ; 17(11)2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27801830

RESUMEN

Checkpoint kinase 2 (CHK2) and cell division cycle 25C (CDC25C) are two proteins involved in the DNA damage response pathway, playing essential roles in maintaining genome integrity. As one of the major hallmarks of abnormal cellular division, genomic instability occurs in most cancers. In this study, we identified the functional expression of pCHK2-Thr68 and pCDC25C-Ser216 in breast cancer, as well as its association with breast cancer survival. Tissue microarray analysis using immunohistochemistry was constructed to identify the expression of pCHK2-Thr68 and pCDC25C-Ser216 in 292 female breast cancer patients. The relationship among protein expression, clinicopathological factors (e.g., human epidermal growth factor receptor 2 (HER 2), tumor size, tumor-node-metastasis (TNM) classification), and overall survival of the breast cancer tissues were analyzed using Pearson's χ-square (χ²) test, Fisher's exact test, multivariate logistic regression and Kaplan-Meier survival analysis. Significantly higher expressions of pCHK2-Thr68 and pCDC25C-Ser216 were observed in the nucleus of the breast cancer cells compared to the paracancerous tissue (pCHK2-Thr68, 20.38% vs. 0%; pCDC25C-Ser216, 82.26% vs. 24.24%). The expression of pCHK2-Thr68 and pCDC25C-Ser216 in breast cancer showed a positive linear correlation (p = 0.026). High expression of pCHK2-Thr68 was associated with decreased patient survival (p = 0.001), but was not an independent prognostic factor. Our results suggest that pCHK2-Thr68 and pCDC25C-Ser216 play important roles in breast cancer and may be potential treatment targets.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias de la Mama/genética , Quinasa de Punto de Control 2/biosíntesis , Fosfatasas cdc25/biosíntesis , Adulto , Anciano , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Quinasa de Punto de Control 2/genética , Daño del ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica/genética , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática/genética , Metástasis Linfática/patología , Análisis por Micromatrices , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Receptor ErbB-2/genética , Fosfatasas cdc25/genética
5.
Mar Drugs ; 14(9)2016 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-27626431

RESUMEN

R-Phycoerythrin (R-PE), one of the chemical constituents of red algae, could produce singlet oxygen upon excitation with the appropriate radiation and possibly be used in photodynamic therapy (PDT) for cancer. Documents reported that R-PE could inhibit cell proliferation in HepG2 and A549 cells, which was significative for cancer therapy. This is due to the fact that R-PE could kill cancer cells directly as well as by PDT. However, little is known about the cytotoxicity of R-PE to the SGC-7901 cell. In this study, it has been found that R-PE could inhibit SGC-7901 proliferation and induce cell apoptosis, which was achieved by arresting the SGC-7901 cell at S phase. CyclinA, CDK2 and CDC25A are proteins associated with the S phase, and it was found that R-PE could increase the expression of cyclin A protein and decrease the expression of CDK2 and CDC25A proteins. Thus, it was concluded that R-PE reduced the CDK2 protein activated through decreasing the CDC25A factor, which reduced the formation of Cyclin-CDK complex. The reduction of Cyclin-CDK complex made the SGC-7901 cells arrest at the S phase. Therefore, R-PE induced apoptosis by arresting the SGC-7901 cell at S phase was successful, which was achieved by the expression of the CDC25A protein, which reduced the CDK2 protein actived and the formation of Cyclin-CDK complex.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ficoeritrina/farmacología , Fase S/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina A/biosíntesis , Ciclina A/genética , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 2 Dependiente de la Ciclina/genética , Humanos , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética
6.
Mol Cell Biochem ; 416(1-2): 33-46, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27038604

RESUMEN

Alterations in the cell-cycle regulatory genes result in uncontrolled cell proliferation leading to several disease conditions. Cyclin-dependent kinases (CDK) and their regulatory subunit, cyclins, are essential proteins in cell-cycle progression. The activity of CDK is regulated by a series of phosphorylation and dephosphorylation at different amino acid residues. Cell Division Cycle-25 (CDC25) plays an important role in transitions between cell-cycle phases by dephosphorylating and activating CDKs. CDC25B and CDC25C play a major role in G2/M progression, whereas CDC25A assists in G1/S transition. Different isomers of CDC25 expressions are upregulated in various clinicopathological situations. Overexpression of CDC25A deregulates G1/S and G2/M events, including the G2 checkpoint. CDC25B has oncogenic properties. Binding to the 14-3-3 proteins regulates the activity and localization of CDC25B. CDC25C is predominantly a nuclear protein in mammalian cells. At the G2/M transition, mitotic activation of CDC25C protein occurs by its dissociation from 14-3-3 proteins along with its phosphorylation at multiple sites within its N-terminal domain. In this article, we critically reviewed the biology of the activation/deactivation of CDC25 by kinases/phosphatases to maintain the level of CDK-cyclin activities and thus the genomic stability, clinical implications due to dysregulation of CDC25, and potential role of CDC25 inhibitors in diseases.


Asunto(s)
Quinasas Ciclina-Dependientes/biosíntesis , Regulación Enzimológica de la Expresión Génica , Inestabilidad Genómica , Meiosis , Fosfatasas cdc25/biosíntesis , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animales , Quinasas Ciclina-Dependientes/genética , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Fosforilación , Fosfatasas cdc25/genética
7.
BMC Cancer ; 16: 246, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27012847

RESUMEN

BACKGROUND: ACFP is an anti-cancer fusion peptide derived from bovine milk protein. This study was to investigate the anti-cancer function and underlying mechanisms of ACFP in ovarian cancer. METHODS: Fresh ovarian tumor tissues were collected from 53 patients who underwent initial debulking surgery, and primary cancer cells were cultured. Normal ovarian surface epithelium cells (NOSECs), isolated from 7 patients who underwent surgery for uterine fibromas, were used as normal control tissue. Anti-viabilities of ACFP were assessed by WST-1 (water-soluble tetrazolium 1), and apoptosis was measured using a flow cytometry-based assay. Gene expression profiles of ovarian cancer cells treated with ACFP were generated by cDNA microarray, and the expression of apoptotic-specific genes, such as bcl-xl, bax, akt, caspase-3, CDC25C and cyclinB1, was assessed by real time PCR and western blot analysis. RESULTS: Treatment with ACFP inhibited the viability and promoted apoptosis of primary ovarian cancer cells but exhibited little or no cytotoxicity toward normal primary ovarian cells. Mechanistically, the anti-cancer effects of ACFP in ovarian cells were shown to occur partially via changes in gene expression and related signal pathways. Gene expression profiling highlighted that ACFP treatment in ovarian cancer cells repressed the expression of bcl-xl, akt, CDC25C and cyclinB1 and promoted the expression of bax and caspase-3 in a time- and dose-dependent manner. CONCLUSIONS: Our results suggest that ACFP may represent a potential therapeutic agent for ovarian cancer that functions by altering the expression and signaling of cancer-related pathways in ovarian cancer cells.


Asunto(s)
Proteínas de la Leche/administración & dosificación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias Ováricas/tratamiento farmacológico , Péptidos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Bovinos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina B1/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/biosíntesis , Fosfatasas cdc25/biosíntesis
8.
Cancer Sci ; 106(10): 1341-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26250568

RESUMEN

Magnolol, the major active compound found in Magnolia officinalis has a wide range of clinical applications due to its anti-inflammation and anti-oxidation effects. This study investigated the effects of magnolol on the growth of human gallbladder carcinoma (GBC) cell lines. The results indicated that magnolol could significantly inhibit the growth of GBC cell lines in a dose- and time-dependent manner. Magnolol also blocked cell cycle progression at G0 /G1 phase and induced mitochondrial-related apoptosis by upregulating p53 and p21 protein levels and by downregulating cyclin D1, CDC25A, and Cdk2 protein levels. When cells were pretreated with a p53 inhibitor (pifithrin-a), followed by magnolol treatment, pifithrin-a blocked magnolol-induced apoptosis and G0 /G1 arrest. In vivo, magnolol suppressed tumor growth and activated the same mechanisms as were activated in vitro. In conclusion, our study is the first to report that magnolol has an inhibitory effect on the growth of GBC cells and that this compound may have potential as a novel therapeutic agent for the treatment of GBC.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/patología , Lignanos/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/biosíntesis , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Medicina Tradicional China , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/biosíntesis
9.
Mol Med Rep ; 12(1): 1465-72, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25816175

RESUMEN

Esophageal cancer is a common malignant tumor worldwide with a high incidence rate in China and it is a great threat to human health. Combined modality therapy is used for chemotherapeutic treatment of esophageal cancer; however, drug resistance and side effects of the drugs is a major barrier to the success of chemotherapy. As chemotherapy with common drugs is far from providing satisfactory clinical outcomes for patients with esophageal cancer, more efficient drugs are urgently required. Artesunate (Art) is the first-line treatment option for malaria; however, it was recently revealed that Art has remarkable anti-tumor activity, making it a novel candidate for cancer chemotherapy. Although the anti-cancer effects of Art have been well documented, its potential against esophageal cancer has rarely been explored. The present study aimed to investigate the significance and mechanism of the anti-proliferative activity of Art on esophageal cancer cells in vitro and in vivo. In the in vitro experiments, Art inhibited the growth as well as induced cell apoptosis and cell cycle arrest of esophageal cancer cell lines (Eca109 and Ec9706) in a concentration-dependent manner. Furthermore, downregulation of mitochondrial membrane potential, B-cell lymphoma-2 (BCL-2) and CDC25A, as well as upregulation of BCL-2­associated X protein (Bax) and caspase-3 expression in Art-treated cells were identified. In addition, an in vivo study showed that Art produced a dose-dependent tumor regression in nude mice, while side effects were low. The anti-tumor activity of 200 mg/kg Art was similar to that of 3 mg/kg cisplatin. In conclusion, Art exerted concentration-dependent inhibitory activity against esophageal cancer in vivo and in vitro by inducing cell apoptosis and cell cycle arrest through affecting mitochondrial membrane potential, BCL-2, Bax, caspase-3 and CDC25A.


Asunto(s)
Artemisininas/administración & dosificación , Cisplatino/administración & dosificación , Neoplasias Esofágicas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Artesunato , Caspasa 3/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/biosíntesis , Fosfatasas cdc25/biosíntesis
10.
Pharmacol Rep ; 67(2): 245-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25712646

RESUMEN

BACKGROUND: Deoxypodophyllotoxin (DPT) is a semi-synthetic compound derived from the extract of Dysosma versipellis (Hance) M. Cheng, one of the most popular Chinese herbal medicines. The present study evaluates the in vitro cytotoxicity of DPT on a wide panel of human cancer cell lines and investigates its molecular mechanism of action on high grade glioma U-87 MG and SF126 cells. METHODS: The growth inhibitory effect of DPT on different types of human cancer cells was measured by the Cell Counting Kit-8 (CCK-8) assay. For the elucidation of the nature of the cellular response to DPT-treatment; flow cytometry-based assays, light and fluorescent microscopy, caspase colorimetric and inhibition assays, and Western blot analysis were performed. RESULTS: Our data show that DPT possesses a potent growth-inhibitory action, with IC50 values in nanomolar ranges. Cell cycle analysis revealed G2/M phase arrest in a dose- and time-dependent manner before cell death occurred. Additional studies indicated that DPT induced G2 arrest in U-87 MG cells by decreasing the expression of Cdc2, cyclin B1, and Cdc25C proteins. In contrast, DPT failed to down-regulate these cell cycle regulatory molecules in SF126 glioblastoma cells and stopped the cell cycle at M phase. Interestingly, morphological changes and biochemical markers such as phosphatydylserine externalization, DNA fragmentation, and caspase activation, confirmed that DPT-treatment resulted in an induction of apoptosis in both examined cell lines via caspase-dependent pathways. CONCLUSIONS: Taken together, our data demonstrated that DPT possesses a potent in vitro cytotoxic activity and exerts its effect via G2/M arrest and apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/patología , Podofilotoxina/análogos & derivados , Proteína Quinasa CDC2 , Caspasas/metabolismo , Recuento de Células , Línea Celular Tumoral , Ciclina B1/biosíntesis , Quinasas Ciclina-Dependientes/biosíntesis , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Podofilotoxina/farmacología , Fosfatasas cdc25/biosíntesis
11.
Tumour Biol ; 36(7): 5001-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25680404

RESUMEN

The non-coding microRNAs (miRNAs) have tissue- and disease-specific expression patterns. Dysregulation of miRNAs has been associated with initiation and progression of oncogenesis in humans. The abnormal expression of CDC25B phosphatases detected in a number of tumors implies that their dysregulation is involved in malignant transformation. Using miRNA target prediction software, we found that miR-211 could target the 3'UTR sequence of CDC25B. To shed light on their roles of miR-211 in breast cancer, the expression of miR-211 was examined by real-time RT-PCR in breast cancer and normal tissues. MiR-211 is significantly downregulated in breast cancer. MiR-211 re-expression suppressed cell growth, cell cycle, migration, and invasion in triple-negative breast cancer (TNBC) cell line MDA-MB231. Luciferase expression from a reporter vector containing the CDC25B -3'UTR was decreased when this construct was transfected with miR-211. The over-expression of miR-211 suppressed the endogenous CDC25B protein level in TNBC cells. For the first time, we demonstrate that miRNA-211 is a direct negative regulator of CDC25B expression in TNBC cells, alters other related target proteins CCNB1 and FOXM1, and then inhibits breast cancer cells growth, migration, and invasion and lead G2/M arrest. The transcriptional loss of miR-211 and the resultant increase in CDC25B expression facilitate increased genomic instability at an early stage of tumor development.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , MicroARNs/genética , Neoplasias de la Mama Triple Negativas/genética , Fosfatasas cdc25/biosíntesis , Línea Celular Tumoral , Ciclina B1/biosíntesis , Ciclina B1/genética , Femenino , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Invasividad Neoplásica/genética , Neoplasias de la Mama Triple Negativas/patología , Fosfatasas cdc25/genética
12.
Oncol Rep ; 33(3): 1434-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25591797

RESUMEN

Infection with Epstein-Barr virus (EBV) and its encoded latent membrane protein 1 (LMP1) play oncogenic roles in Burkitt's lymphoma (BL). Flow cytometry was used to measure cellular reactive oxygen species (ROS) concentrations, and cellular lactate generation and diphenylene iodonium (DPI) cytotoxicity were determined by analyzing lactate concentrations and cell viability. We also measured NAD(P)H oxidase (NOX) activity. Reverse transcriptase PCR and qPCR assays were used to analyze LMP1 levels, and protein expression was measured by immunoblotting. In the present study, EBV was able to induce NOX activity and ROS generation in the BL cells. Inhibition of NOX activity by DPI suppressed ROS levels and elevated lactate levels. DPI treatment first resulted in a G2-M phase cell cycle arrest and then induced significant apoptosis. Immunoblot analysis demonstrated that DPI suppressed the expression of c-Myc and Cdc25A within 6 h, which may have caused the cell cycle arrest. Collectively, these findings indicate a close relationship between EBV infection and NOX activation, permitting a deeper understanding of ROS inhibition in cell cycle regulation and providing a novel therapeutic target for BL treatment.


Asunto(s)
Linfoma de Burkitt/patología , Herpesvirus Humano 4/patogenicidad , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas de la Matriz Viral/metabolismo , Apoptosis/efectos de los fármacos , Linfoma de Burkitt/virología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Citometría de Flujo , Herpesvirus Humano 4/genética , Humanos , Ácido Láctico/biosíntesis , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , NADPH Oxidasas/biosíntesis , Compuestos Onio/farmacología , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de la Matriz Viral/genética , Fosfatasas cdc25/biosíntesis
13.
Int J Oncol ; 46(2): 578-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25384584

RESUMEN

We evaluated the cytostatic effect of 6,7-di-O-acetyl-sinococuline (FK-3000) isolated from Stephania delavayi Diels. against breast carcinoma cell lines MDA-MB­231 and MCF-7. FK-3000 suppressed CDC25B phosphorylation directly and indirectly via p38 MAPK phosphorylation. CDC25B dephosphorylation decreased levels of cyclin B and phospho-CDC-2, and ultimately induced cell cycle arrest at the G2/M phase. The p38 MAPK inhibitor, SB 239063 blocked FK-3000-induced p38 MAPK phosphorylation and nuclear accumulation, but did not completely rescue cell death. Conclusively FK-3000 exerts its antiproliferative effect through two pathways: i) G2/M cell cycle arrest via downregulation of cyclin B and phospho-CDC2 by p38 MAPK phosphorylation and CDC25B dephosphorylation, and ii) p38 MAPK-independent induction of apoptosis.


Asunto(s)
Alcaloides/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Fosfatasas cdc25/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma/genética , Carcinoma/patología , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Int J Oncol ; 45(6): 2393-402, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25340978

RESUMEN

Cantharidin (CTD), a component of natural mylabris (Mylabris phalerata Pallas) was reported to have high cytotoxicity in many human cancer cell lines. However, it was not reported to affect human melanoma A375.S2 cells. In the present study, we found that CTD induced cell morphological changes and decreased the percentage of viable cells and induced G2/M phase arrest and induction of apoptosis in A375.S2 cells. Results also showed that CTD induced the generation of reactive oxygen species (ROS) and Ca2+ and decreased mitochondria membrane potential and lead to the release of cytochrome c, AIF and Endo G. Further investigation revealed that CTD induced A375.S2 cells with an increase of caspase activation and caspase-dependent apoptotic proteins to trigger correlated pathway mechanisms according to western blotting results. Western blotting was used for examining the changes of G2/M phase arrest and apoptosis-associated protein expression and confocal laser microscopy was used to examine the translocation apoptosis-associated protein. Results showed that CTD increased the protein expression of caspase-3, -8 and -9, cytochrome c, Bax, Bid, Endo G and AIF but inhibited the levels of Bcl-2 and Bcl-x. CTD induced ER stress-associated protein expression such as GRP78, IRE1ß, ATF6α and caspase-12. Based on those observations, we suggest that CTD may have potential as a novel anti-cancer agent for the treatment of skin cancer.


Asunto(s)
Cantaridina/administración & dosificación , Ciclina A/biosíntesis , Melanoma/tratamiento farmacológico , Fosfatasas cdc25/biosíntesis , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ciclina A/antagonistas & inhibidores , Chaperón BiP del Retículo Endoplásmico , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/genética , Melanoma/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Neoplasias/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Fosfatasas cdc25/antagonistas & inhibidores
15.
Mol Cell Biol ; 34(23): 4216-31, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25266660

RESUMEN

Recently, we demonstrated that the microRNA 424(322)/503 [miR-424(322)/503] cluster is transcriptionally controlled by transforming growth factor ß (TGF-ß) in the mammary epithelium. Induction of this microRNA cluster impacts mammary epithelium fate by regulating apoptosis and insulin-like growth factor 1 (IGF1) signaling. Here, we expanded our finding to demonstrate that miR-424(322)/503 is an integral component of the cell cycle arrest mediated by TGF-ß. Mechanistically, we showed that after TGF-ß exposure, increased levels of miR-424(322)/503 reduce the expression of the cell cycle regulator CDC25A. miR-424(322)/503-dependent posttranscriptional downregulation of CDC25A cooperates with previously described transcriptional repression of the CDC25A promoter and proteasome-mediated degradation to reduce the levels of CDC25A expression and to induce cell cycle arrest. We also provide evidence that the TGF-ß/miR-424(322)/503 axis is part of the mechanism that regulates the proliferation of hormone receptor-positive (HR(+)) mammary epithelial cells in vivo.


Asunto(s)
Glándulas Mamarias Humanas/crecimiento & desarrollo , MicroARNs/genética , Factor de Crecimiento Transformador beta/metabolismo , Fosfatasas cdc25/biosíntesis , Animales , Apoptosis/genética , Línea Celular , Proliferación Celular/genética , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/biosíntesis , Regiones Promotoras Genéticas , Pirazoles/farmacología , Pirroles/farmacología , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Fosfatasas cdc25/genética
16.
Asian Pac J Cancer Prev ; 15(7): 3293-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24815485

RESUMEN

BACKGROUND: Pine needle oil from crude extract of pine needles has anti-tumor effects, but the effective component is not known. METHODS: In the present study, compounds from a steam distillation extract of pine needles were isolated and characterized. Alpha-pinene was identified as an active anti-proliferative compound on hepatoma carcinoma BEL-7402 cells using the MTT assay. RESULTS: Further experiments showed that α-pinene inhibited BEL-7402 cells by arresting cell growth in the G2/M phase of the cell cycle, downregulating Cdc25C mRNA and protein expression, and reducing cycle dependence on kinase 1(CDK1) activity. CONCLUSION: Taken together, these findings indicate that α-pinene may be useful as a potential anti-tumor drug.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Monoterpenos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Monoterpenos Bicíclicos , Proteína Quinasa CDC2 , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/biosíntesis , Regulación hacia Abajo , Humanos , Pinus/metabolismo , Extractos Vegetales/farmacología , ARN Mensajero/biosíntesis , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética
17.
Tumour Biol ; 35(3): 2503-12, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24234332

RESUMEN

Cdc25 dual-specicity phosphatases are essential regulators at critical stages of cell cycle. Cdc25B is overexpressed in several human tumor types. The activity of Cdc25B is regulated by 14-3-3 dimer. To investigate the roles of Cdc25B and 14-3-3σ in bladder carcinoma, we examined expressions of Cdc25B and 14-3-3σ proteins in bladder carcinoma and cell lines and analyzed their roles in the development and prognosis of urinary bladder carcinoma. Immunohistochmistry was used to detect the expressions of Cdc25B and 14-3-3σ in 105 bladder carcinomas. Moreover, expressions of Cdc25B and 14-3-3σ were analyzed by real-time PCR and Western blot in 40 bladder carcinomas and 20 normal epithelial tissues. Specific siRNA was used to knockdown the expression of Cdc25B or 14-3-3σ. Wild-type plasmid was used to overexpress 14-3-3σ. MTT assay and Flow cytometry were used to examine proliferation and cell cycle of bladder cancer cells. There were higher Cdc25B expression and lower 14-3-3σ expression in carcinomas than in the adjacent normal tissues (P < 0.05), positive and negative correlations being noted with clinical stage and histopathologic grade. Cdc25B expression was positively correlated with recurrence and poor prognosis. Downregulation of Cdc25B resulted in slower growth, more G2/M cells and 14-3-3σ increasing. However, upregulation and downregulation of 14-3-3σ did not affect cell growth and Cdc25B expression. It showed that Cdc25B upregulation and 14-3-3σ downregulation might promote development of bladder cancer and suggested a poor prognosis. Moreover, Cdc25B could play an important role on the bladder cancer cell proliferation and cell cycle progression and regulate expression of 14-3-3σ.


Asunto(s)
Proteínas 14-3-3/biosíntesis , Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Transicionales/metabolismo , Exorribonucleasas/biosíntesis , Neoplasias de la Vejiga Urinaria/metabolismo , Fosfatasas cdc25/biosíntesis , Biomarcadores de Tumor/análisis , Western Blotting , Carcinoma de Células Transicionales/mortalidad , Carcinoma de Células Transicionales/patología , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología
18.
Am J Pathol ; 184(1): 110-21, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24211536

RESUMEN

Hepatic cystogenesis in polycystic liver diseases is associated with abnormalities of cholangiocyte cilia. Given the crucial association between cilia and centrosomes, we tested the hypothesis that centrosomal defects occur in cystic cholangiocytes of rodents (Pkd2(WS25/-) mice and PCK rats) and of patients with polycystic liver diseases, contributing to disturbed ciliogenesis and cyst formation. We examined centrosomal cytoarchitecture in control and cystic cholangiocytes, the effects of centrosomal abnormalities on ciliogenesis, and the role of the cell-cycle regulator Cdc25A in centrosomal defects by depleting cholangiocytes of Cdc25A in vitro and in vivo and evaluating centrosome morphology, cell-cycle progression, proliferation, ciliogenesis, and cystogenesis. The cystic cholangiocytes had atypical centrosome positioning, supernumerary centrosomes, multipolar spindles, and extra cilia. Structurally aberrant cilia were present in cystic cholangiocytes during ciliogenesis. Depletion of Cdc25A resulted in i) a decreased number of centrosomes and multiciliated cholangiocytes, ii) an increased fraction of ciliated cholangiocytes with longer cilia, iii) a decreased proportion of cholangiocytes in G1/G0 and S phases of the cell cycle, iv) decreased cell proliferation, and v) reduced cyst growth in vitro and in vivo. Our data support the hypothesis that centrosomal abnormalities in cholangiocytes are associated with aberrant ciliogenesis and that accelerated cystogenesis is likely due to overexpression of Cdc25A, providing additional evidence that pharmacological targeting of Cdc25A has therapeutic potential in polycystic liver diseases.


Asunto(s)
Centrosoma/metabolismo , Centrosoma/ultraestructura , Quistes/metabolismo , Quistes/ultraestructura , Hepatopatías/metabolismo , Fosfatasas cdc25/biosíntesis , Animales , Conductos Biliares/patología , Western Blotting , Cilios/metabolismo , Cilios/ultraestructura , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Humanos , Ratones , Microscopía Confocal , Microscopía Electrónica , Ratas
19.
Int J Cancer ; 134(8): 1991-2002, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24150948

RESUMEN

Targeting the cancer cell cycle machinery is an important strategy for cancer treatment. Cdc25A is an essential regulator of cycle progression and checkpoint response. Over-expression of Cdc25A occurs often in human cancers. In this study, we show that Rocaglamide-A (Roc-A), a natural anticancer compound isolated from the medicinal plant Aglaia, induces a rapid phosphorylation of Cdc25A and its subsequent degradation and, thereby, blocks cell cycle progression of tumor cells at the G1-S phase. Roc-A has previously been shown to inhibit tumor proliferation by blocking protein synthesis. In this study, we demonstrate that besides the translation inhibition Roc-A can induce a rapid degradation of Cdc25A by activation of the ATM/ATR-Chk1/Chk2 checkpoint pathway. However, Roc-A has no influence on cell cycle progression in proliferating normal T lymphocytes. Investigation of the molecular basis of tumor selectivity of Roc-A by a time-resolved microarray analysis of leukemic vs. proliferating normal T lymphocytes revealed that Roc-A activates different sets of genes in tumor cells compared with normal cells. In particular, Roc-A selectively stimulates a set of genes responsive to DNA replication stress in leukemic but not in normal T lymphocytes. These findings further support the development of Rocaglamide for antitumor therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Benzofuranos/farmacología , Quinasa de Punto de Control 2/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/genética , Daño del ADN/efectos de los fármacos , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Células Jurkat , Leucemia/tratamiento farmacológico , Células MCF-7 , Fosforilación/efectos de los fármacos , Extractos Vegetales/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Fosfatasas cdc25/biosíntesis , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
20.
Oncol Res ; 22(3): 147-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-26168133

RESUMEN

Berberine is a clinically important natural isoquinoline alkaloid found in many medicinal herbs. Berberine has been shown to have many pharmacological effects including antimicrobial, antitumor, and anti-inflammatory activities. However, the effects and mechanism of action of berberine have not been studied in chondrosarcoma. Therefore, the effects of berberine on proliferation in a human chondrosarcoma cell line (HTB-94) were investigated. Berberine inhibited cell proliferation in a concentration-dependent manner. We also determined that inhibition of cell proliferation by berberine occurred via G2/M phase arrest in HTB-94 cells. Berberine induced cell cycle arrest at the G2/M phase by upregulation of p53 and p21 expression and suppressed cyclin B1, cyclin-dependent kinase 1 (cdc2), cdc25c, and phosphorylated retinoblastoma tumor-suppressor protein (pRb) expression. In addition, berberine stimulated phosphorylation of protein kinase B (Akt) and p38 kinase. Inhibition of phosphatidylinositol 3-kinase (PI3K)/Akt with LY294002 (LY) and p38 kinase with SB203580 (SB), respectively, decreased berberine-induced p53 and p21 expression and restored cell proliferation and expression of cyclin B1, cdc2, cdc25c, and pRb cell cycle progression proteins. These results suggest that berberine-induced inhibition of cell proliferation by cell cycle arrest at the G2/M phases was regulated through PI3K/Akt and p38 kinase pathways in HTB-94 chondrosarcoma cells.


Asunto(s)
Berberina/farmacología , Proliferación Celular/efectos de los fármacos , Condrosarcoma/genética , Activación Enzimática/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Proteína Quinasa CDC2 , Línea Celular Tumoral , Condrosarcoma/tratamiento farmacológico , Cromonas/farmacología , Ciclina B1/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Quinasas Ciclina-Dependientes/biosíntesis , Humanos , Imidazoles/farmacología , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Fosfatasas cdc25/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA