Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Asian Pac J Cancer Prev ; 23(1): 151-160, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35092383

RESUMEN

BACKGROUND: Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death in females worldwide. Schleichera oleosa (kusum tree) belongs to the Sapindaceae family commonly found in many states of India. This plant is traditionally being used in various pathological conditions. METHODS: In vitro studies were performed using seed extract of Schleichera oleosa. Different concentrations of seed extracts were treated on MCF-7 breast cancer cell line and its effect on migration and colony formation were observed. BRCA1 and p16 gene expression was analyzed by real-time PCR and Western blotting. RESULTS: We have analyzed anticancer and anti-metastatic effects of seed extract in breast cancer and IC50 was 140µg/ml concentration. Further, its inhibitory role in cell migration and colony formation was at 140µg/ml (P<0.0001) concentration and reduced significantly growth of sphere at 140 µg (P<0.0031) and 150µg (P<0.0010) concentration after 5 days of treatment. The apoptosis study was shown a significant increase at 140 µg (P<0.0001) in apoptotic cells. Expression of BRCA1 and p16 were found to be over-expressed as 1.4 and 1.7 fold, respectively, at 140µg/ml concentration after 24 h of treatment at the transcription level. BRCA1 protein was up-regulated but p16 expression down-regulated at 140 to 150µg/ml (One-Way ANOVA, P<0.0001) concentration. CONCLUSION: In this study, we found a significant role of S. Oleosa seed extract has an anti-cancer as well as anti-metastatic via up-regulation of BRCA1 and p16 genes in breast cancer cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Genes BRCA1/efectos de los fármacos , Genes p16/efectos de los fármacos , Extractos Vegetales/farmacología , Sapindaceae , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Semillas , Regulación hacia Arriba
2.
Clin Exp Dermatol ; 46(8): 1551-1554, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34115902

RESUMEN

Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine neoplasm of the skin, which has an exceedingly poor prognosis. The AKT/mammalian target of rapamycin (mTOR) signalling pathway, which plays a pivotal role in the modulation of protein synthesis and cell survival, has been shown to be extremely important for Merkel cell carcinogenesis. In the current study, we found that AKT has important regulatory functions in MCC cells and that inhibition of AKT with the novel ATP-competitive AKT inhibitor, afuresertib, has widespread effects on proliferative pathways. In particular, we found that treatment of MCC cells with afuresertib led to deactivation of mTOR and glycogen synthase kinase 3 pathway proteins while increasing activation of proapoptotic pathways through the upregulation of p16 expression and phosphomodulation of the B-cell lymphoma-2-associated death promoter. Overall, afuresertib treatment led to significant and robust inhibition of MCC cell proliferation, thus raising intriguing questions regarding the potential efficacy of AKT inhibition for the future clinical management of MCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células de Merkel/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirazoles/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Tiofenos/uso terapéutico , Transactivadores/antagonistas & inhibidores , Carcinoma de Células de Merkel/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Genes p16/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Neoplasias Cutáneas/patología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba
3.
Biochem Pharmacol ; 183: 114320, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33161023

RESUMEN

Cell cycle control is often disrupted in gastric cancer (GC), making it an attractive therapeutic target. Abemaciclib is a specific CDK4/6 inhibitor that has been shown to improve treatment efficacy in hormone receptor-positive advanced breast cancer; however, its potential therapeutic value and predictive markers have not yet been revealed in GC. In this study, we investigated the efficacy of abemaciclib using preclinical GC models representing defined molecular subtypes from The Cancer Genome Atlas. In these 49 GC cell lines, Epstein-Barr virus (EBV) and high microsatellite instability (MSI-H)-type cell lines were p16 methylated and sensitive to abemaciclib; further, genomically stable (GS), and chromosomal instability (CIN)-type cell lines with p16 methylation and intact Rb were also found to be responsive. In addition, we found that GC patients with p16 methylation often displayed a poor prognosis. Collectively, these data provide a foundation for clinical trials to assess the therapeutic efficacy of abemaciclib in GC and suggest that p16 methylation could be used as a predictive marker to identify patients with GC who may benefit from abemaciclib-based therapies.


Asunto(s)
Aminopiridinas/farmacología , Bencimidazoles/farmacología , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Genes p16/efectos de los fármacos , Neoplasias Gástricas/metabolismo , Aminopiridinas/uso terapéutico , Animales , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Metilación de ADN/fisiología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/fisiología , Femenino , Genes p16/fisiología , Humanos , Ratones , Ratones Desnudos , Valor Predictivo de las Pruebas , Neoplasias Gástricas/tratamiento farmacológico
4.
Aging Cell ; 19(1): e13060, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31773901

RESUMEN

Although aging and senescence have been extensively studied in the past few decades, however, there is lack of clinical treatment available for anti-aging. This study presents the effects of berberine (BBR) on the aging process resulting in a promising extension of lifespan in model organisms. BBR extended the replicative lifespan, improved the morphology, and boosted rejuvenation markers of replicative senescence in human fetal lung diploid fibroblasts (2BS and WI38). BBR also rescued senescent cells with late population doubling (PD). Furthermore, the senescence-associated ß-galactosidase (SA-ß-gal)-positive cell rates of late PD cells grown in the BBR-containing medium were ~72% lower than those of control cells, and its morphology resembled that of young cells. Mechanistically, BBR improved cell growth and proliferation by promoting entry of cell cycles from the G0 or G1 phase to S/G2 -M phase. Most importantly, BBR extended the lifespan of chemotherapy-treated mice and naturally aged mice by ~52% and ~16.49%, respectively. The residual lifespan of the naturally aged mice was extended by 80%, from 85.5 days to 154 days. The oral administration of BBR in mice resulted in significantly improved health span, fur density, and behavioral activity. Therefore, BBR may be an ideal candidate for the development of an anti-aging medicine.


Asunto(s)
Berberina/uso terapéutico , Ciclinas/metabolismo , Genes p16/efectos de los fármacos , Medicina Tradicional China/métodos , Animales , Berberina/farmacología , Senescencia Celular , Humanos , Masculino , Ratones
5.
Toxicol Ind Health ; 35(6): 431-444, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31131716

RESUMEN

Man-made mineral fibers (MMMFs) are substitutes for asbestos. MMMFs are widely used as insulation, but their molecular mechanisms underlying the tumorigenic effects in vivo have been poorly studied. For this reason, this work aimed to explore the properties and carcinogenic molecular mechanisms of MMMFs. The three MMMFs, rock wool (RW), glass fibers (GFs), and ceramic fibers (CFs), were prepared into respirable dust. Particle size, morphology, and chemical composition were analyzed by laser particle analyzer, scanning electron microscope, and X-ray fluorescence spectrometer, respectively. The Wistar rats were administered multiple intratracheal instillations of three MMMFs once a month. Then, several parameters (e.g. body mass, lung mass, and lung histology) were measured at 1, 3, and 6 months. After that, levels of P53, P16, C-JUN, and C-FOS mRNA and protein were measured by quantitative real-time reverse transcription polymerase chain reaction and Western blotting. This work found that exposure to MMMFs could influence the growth of body mass and increase lung mass. General conditions showed white nodules and irregular atrophy. In addition, MMMFs could lead to inactivation of anti-oncogene P16 and activation of proto-oncogenes (C-JUN and C-FOS) in the mRNA and protein levels, in which GF and CF were more obvious compared with RW. The effect of MMMFs was different, which may be related to the physical and chemical characteristics of different MMMFs. In conclusion, MMMFs (GF and CF) could induce an unbalanced expression of cancer-related genes in the lung tissues of rats. The understanding of the determinants of toxicity and carcinogenicity provides a scientific basis for developing and introducing new safer MMMF products, and the present study provides some useful insights into the carcinogenic mechanism of MMMFs.


Asunto(s)
Lesión Pulmonar/inducido químicamente , Fibras Minerales/toxicidad , Oncogenes/efectos de los fármacos , Animales , Genes fos/efectos de los fármacos , Genes jun/efectos de los fármacos , Genes p16/efectos de los fármacos , Genes p53/efectos de los fármacos , Pulmón , Lesión Pulmonar/patología , Masculino , ARN Mensajero/análisis , Ratas , Ratas Wistar
6.
Sci Rep ; 8(1): 7494, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29748642

RESUMEN

Endocrine disrupting chemicals (EDC) interfere with the natural hormone balance and may induce epigenetic changes through exposure during sensitive periods of development. In this study, the effects of short-term estradiol-17ß (E2) exposure on various tissues of pregnant sows (F0) and on day 10 blastocysts (F1) were assessed. Intergenerational effects were investigated in the liver of 1-year old female offspring (F1). During gestation, sows were orally exposed to two low doses and a high dose of E2 (0.05, 10, and 1000 µg/kg body weight/day). In F0, perturbed tissue specific mRNA expression of cell cycle regulation and tumour suppressor genes was found at low and high dose exposure, being most pronounced in the endometrium and corpus luteum. The liver showed the most significant DNA hypomethylation in three target genes; CDKN2D, PSAT1, and RASSF1. For CDKN2D and PSAT1, differential methylation in blastocysts was similar as observed in the F0 liver. Whereas blastocysts showed hypomethylation, the liver of 1-year old offspring showed subtle, but significant hypermethylation. We show that the level of effect of estrogenic EDC, with the periconceptual period as a sensitive time window, is at much lower concentration than currently presumed and propose epigenetics as a sensitive novel risk assessment parameter.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Estradiol/administración & dosificación , Estradiol/efectos adversos , Genes p16/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Transaminasas/genética , Administración Oral , Animales , Cálculo de Dosificación de Drogas , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Disruptores Endocrinos/administración & dosificación , Disruptores Endocrinos/efectos adversos , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes cdc/efectos de los fármacos , Exposición Materna/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Porcinos/embriología , Porcinos/crecimiento & desarrollo
7.
Toxicology ; 400-401: 28-39, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29567467

RESUMEN

T-2 toxin is a worldwide trichothecenetoxin and can cause various toxicities.T-2 toxin is involved in G1 phase arrest in several cell lines but molecular mechanism is still not clear. In present study, we used rat pituitary GH3 cells to investigate the mechanism involved in cell cycle arrest against T-2 toxin (40 nM) for 12, 24, 36 and 48 h as compared to control cells. GH3 cells showed a considerable increase in reactive oxygen species (ROS) as well as loss in mitochondrial membrane potential (△Ym) upon exposure to the T-2 toxin. Flow cytometry showed a significant time-dependent increase in percentage of apoptotic cells and gel electrophoresis showed the hallmark of apoptosis oligonucleosomal DNA fragmentation. Additionally, T-2 toxin-induced oxidative stress and DNA damage with a time-dependent significant increased expression of p53 favors the apoptotic process by the activation of caspase-3 in T-2 toxin treated cells. Cell cycle analysis by flow cytometry revealed a time-dependent increase ofG1 cell population along with the significant time-dependent up-regulation of mRNA and protein expression of p16 and p21 and significant down-regulation of cyclin D1, CDK4, and p-RB levels further verify the G1 phase arrest in GH3 cells. Morphology of GH3 cells by TEM clearly showed the damage and dysfunction to mitochondria and the cell nucleus. These findings for the first time demonstrate that T-2 toxin induces G1 phase cell cycle arrest by the involvement of p16/Rb pathway, along with ROS mediated oxidative stress and DNA damage with p53 and caspase cascade interaction, resulting in apoptosis in GH3 cells.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Genes p16/efectos de los fármacos , Hipófisis/efectos de los fármacos , Proteína de Retinoblastoma/biosíntesis , Transducción de Señal/efectos de los fármacos , Toxina T-2/toxicidad , Animales , Ciclo Celular/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Genes p16/fisiología , Hipófisis/metabolismo , Hipófisis/ultraestructura , Ratas , Transducción de Señal/fisiología
8.
Medicina (Kaunas) ; 52(5): 298-306, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27746118

RESUMEN

BACKGROUND AND AIM: Resistance to chemotherapy is the key obstacle to the effective treatment of various cancers. Accumulating evidence suggests significant involvement of the epithelial-to-mesenchymal transition (EMT) in the chemoresistance of most cancer types. This study aimed at analyzing the gene expression profile of doxorubicin (DOX)-resistant colorectal cancer cells CX-1. MATERIALS AND METHODS: DOX-resistant CX-1 cell sublines were acquired by stepwise increment of DOX concentrations in cell growth media. Global gene expression profiling was performed using human gene expression microarrays. The expression levels of individual genes were assessed by means of quantitative PCR (qPCR), while the DNA methylation pattern of several selected genes was determined by methylation-specific PCR. RESULTS: Four DOX-resistant CX-1 sublines were established as a valuable tool for cell chemoresistance studies. Altered expression of the EMT, cell adhesion and motility, and chemoresistance-related genes was observed in DOX-resistant cells by genome-wide gene expression analysis. Besides, early and significant upregulation of the key EMT genes ZEB1 (5.8×; P<0.001) and CDH2 (6.2×; P=0.044) was identified by qPCR, with subsequent activation of drug transporter gene ABCC1 (3.3×; P=0.007) and cell stemness gene NANOG (2.4×; P=0.008). Downregulation of TET1 (2.1×; P=0.041) and changes in the methylation status of the p16 gene were also involved in the acquisition of cell resistance to DOX. CONCLUSION: The results of our study suggest possible involvement of the key EMT and drug transporter genes in the early phase of cancer cell chemoresistance development.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias Colorrectales/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Inhibidores de Topoisomerasa II/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Metilación de ADN , Epigénesis Genética , Perfilación de la Expresión Génica , Genes p16/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
9.
Oncol Res ; 23(5): 237-48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27098147

RESUMEN

Raltitrexed (RTX) is an antimetabolite drug used as a chemotherapeutic agent for treating colorectal cancer, malignant mesothelioma, and gastric cancer. The antitumor capacity of RTX is attributed to its inhibitory activity on thymidylate synthase (TS), a key enzyme in the synthesis of DNA precursors. The current study is aimed at investigating the potential antitumor effects of RTX in liver cancer. Using the HepG2 cell line as an in vitro model of liver cancer, we evaluated the effects of RTX on cell proliferation employing both a WST-8 assay and a clone formation efficiency assay. In addition, we monitored the ultrastructure changes of HepG2 cells in response to RTX with transmission electric microscopy. To investigate the mechanism underlying the regulation of cell proliferation by RTX, we analyzed cell cycle using cell flow cytometry. Moreover, real-time PCR and Western blot analyses were conducted to examine expression levels of cell cycle regulatory proteins cyclin A and cyclin-dependent kinase 2 (CDK2), as well as their mediators tumor suppressor genes p53 and p16. Our results demonstrate that RTX inhibits HepG2 proliferation by arresting the cell cycle at G0/G1. This cell cycle arrest function was mediated via downregulation of cyclin A and CDK2. The observed elevated expression of p53 and p16 by RTX may contribute to the reduction of cyclin A/CDK2. Our study indicates that RTX could serve as a potential chemotherapeutic agent in the treatment of hepatocellular carcinoma.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Quinazolinas/farmacología , Tiofenos/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes p16/efectos de los fármacos , Genes p53/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase S/efectos de los fármacos
10.
Int J Oncol ; 46(3): 1317-27, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25585641

RESUMEN

Heme oxygenase-1 was reported previously as a resistance target on acute myelocytic leukemia (AML). We found that HO-1 was resistant to 5-azacytidine (AZA) treatment of myelodysplastic syndrome (MDS), and explored further the relative mechanisms. Patient bone marrow mononuclear cells (n=48) diagnosed as different levels of MDS were collected. Cell growth was evaluated by MTT assay; cell cycle and apoptosis were detected by flow cytometry; mRNA expression was assessed by real-time PCR, protein expression was analyzed through western blotting. Methylation was assessed by MSP. The survival time, and weight of mice were recorded. HO-1 overexpression was observed in SKM-1 cells after AZA treatment comparing to other cell lines. The HO-1 expression in MDS patients with high-risk was higher than in low-risk patients. After HO-1 was silenced by lentivirus-mediated siRNA, the proliferation of SKM-1 cells was effectively inhibited by low concentration AZA, and the cell cycle was arrested in the G0/G1 phase. Upregulation of p16 and changing of p16-relative cell cycle protein was observed after silencing HO-1 in AZA treated SKM-1 cells. In addition, DNMT1 was downregulated following the decrease of HO-1 expression. In vivo, silencing HO-1 inhibited SKM-1 cell growth induced by AZA in a NOD/SCID mouse model. Silencing HO-1 sensitized SKM-1 cells toward AZA, which may be attributed to the influence of HO-1 on AZA-induced p16 demethylation. HO-1 may be one of the targets that enhance the therapeutic effects of AZA on MDS malignant transformation inspiring new treatment methods for high-risk and very high-risk MDS patients in clinical practice.


Asunto(s)
Apoptosis/efectos de los fármacos , Azacitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Genes p16 , Hemo-Oxigenasa 1/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Adulto , Anciano , Animales , Apoptosis/genética , Azacitidina/administración & dosificación , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Femenino , Silenciador del Gen/fisiología , Genes p16/efectos de los fármacos , Células HEK293 , Hemo-Oxigenasa 1/antagonistas & inhibidores , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , ARN Interferente Pequeño/administración & dosificación , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
11.
PLoS One ; 8(8): e71031, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940686

RESUMEN

Hexavalent chromium [Cr(IV)], a well-known industrial waste product and an environmental pollutant, is recognized as a human carcinogen. But its mechanisms of carcinogenicity remain unclear, and recent studies suggest that DNA methylation may play an important role in the carcinogenesis of Cr(IV). The aim of our study was to investigate the effects of Cr(IV) on cell cycle progress, global DNA methylation, and DNA methylation of p16 gene. A human B lymphoblastoid cell line and a human lung cell line A549 were exposed to 5-15 µM potassium dichromate or 1.25-5 µg/cm² lead chromate for 2-24 hours. Cell cycle was arrested at G1 phase by both compounds in 24 hours exposure group, but global hypomethylation occurred earlier than cell cycle arrest, and the hypomethylation status maintained for more than 20 hours. The mRNA expression of p16 was significantly up-regulated by Cr(IV), especially by potassium dichromate, and the mRNA expression of cyclin-dependent kinases (CDK4 and CDK6) was significantly down-regulated. But protein expression analysis showed very little change of p16 gene. Both qualitative and quantitative results showed that DNA methylation status of p16 remained unchanged. Collectively, our data suggested that global hypomethylation was possibly responsible for Cr(IV)-induced G1 phase arrest, but DNA methylation might not be related to up-regulation of p16 gene by Cr(IV).


Asunto(s)
Carcinógenos Ambientales/toxicidad , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Cromo/toxicidad , Metilación de ADN/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes p16/efectos de los fármacos , Humanos , Regiones Promotoras Genéticas/efectos de los fármacos , Solubilidad
12.
Braz J Med Biol Res ; 46(8): 643-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23903687

RESUMEN

MP [4-(3',3'-dimethylallyloxy)-5-methyl-6-methoxyphthalide] was obtained from liquid culture of Pestalotiopsis photiniae isolated from the Chinese Podocarpaceae plant Podocarpus macrophyllus. MP significantly inhibited the proliferation of HeLa tumor cell lines. After treatment with MP, characteristic apoptotic features such as DNA fragmentation and chromatin condensation were observed in DAPI-stained HeLa cells. Flow cytometry showed that MP induced G1 cell cycle arrest and apoptosis in a dose-dependent manner. Western blotting and real-time reverse transcription-polymerase chain reaction were used to investigate protein and mRNA expression. MP caused significant cell cycle arrest by upregulating the cyclin-dependent kinase inhibitor p27(KIP1) protein and p21(CIP1) mRNA levels in HeLa cells. The expression of p73 protein was increased after treatment with various MP concentrations. mRNA expression of the cell cycle-related genes, p21(CIP1), p16(INK4a) and Gadd45α, was significantly upregulated and mRNA levels demonstrated significantly increased translation of p73, JunB, FKHR, and Bim. The results indicate that MP may be a potential treatment for cervical cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzofuranos/administración & dosificación , Endófitos/química , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Xylariales/química , Proteínas Reguladoras de la Apoptosis/genética , Benzofuranos/aislamiento & purificación , Proteínas de Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cycadopsida , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Proteínas de Unión al ADN/efectos de los fármacos , Citometría de Flujo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/efectos de los fármacos , Genes p16/efectos de los fármacos , Células HeLa , Humanos , Proteínas Nucleares/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/efectos de los fármacos , Transcripción Genética , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/efectos de los fármacos
13.
Metallomics ; 4(11): 1167-75, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23073540

RESUMEN

Arsenic is carcinogenic, possibly partly through epigenetic mechanisms. We evaluated the effects of arsenic exposure and metabolism on DNA methylation. Arsenic exposure and methylation efficiency in 202 women in the Argentinean Andes were assessed from concentrations of arsenic metabolites in urine (inorganic arsenic, methylarsonic acid [MMA], and dimethylarsinic acid [DMA]), measured by HPLC-ICPMS. Methylation of CpGs of the tumor suppressor gene p16, the DNA repair gene MLH1, and the repetitive elements LINE1 was measured by PCR pyrosequencing of blood DNA. Genotyping (N = 172) for AS3MT was performed using Sequenom™, and gene expression (N = 90) using Illumina DirectHyb HumanHT-12 v3.0. Median arsenic concentration in urine was 230 µg L(-1) (range 10.1-1251). In linear regression analysis, log(2)-transformed urinary arsenic concentrations were positively associated with methylation of p16 (ß = 0.14, P = 0.0028) and MLH1 (ß = 0.28, P = 0.0011), but not with LINE1. Arsenic concentrations were of borderline significance negatively correlated with expression of p16 (r(s) = -0.20; P = 0.066)), but not with MLH1. The fraction of inorganic arsenic was positively (ß = 0.026; P = 0.010) and DMA was negatively (ß = -0.017, P = 0.043) associated with p16 methylation with no effect of MMA. Carriers of the slow-metabolizing AS3MT haplotype were associated with more p16 methylation (P = 0.022). Arsenic exposure was correlated with increased methylation, in blood, of genes encoding enzymes that suppress carcinogenesis, and the arsenic metabolism efficiency modified the degree of epigenetic alterations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Intoxicación por Arsénico/genética , Arsénico/análisis , Metilación de ADN/efectos de los fármacos , Exposición a Riesgos Ambientales/análisis , Genes p16/efectos de los fármacos , Proteínas Nucleares/genética , Adolescente , Adulto , Argentina , Intoxicación por Arsénico/sangre , Intoxicación por Arsénico/metabolismo , Intoxicación por Arsénico/orina , Femenino , Marcadores Genéticos/genética , Haplotipos , Humanos , Metiltransferasas/genética , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Estadísticas no Paramétricas , Abastecimiento de Agua
14.
Hematology ; 17(1): 41-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22549447

RESUMEN

BACKGROUND: Azacytidine (Aza) was the first demethylation agent identified that may inhibit DNA methyltransferases and reverse DNA hypermethylation, restoring the expression of silenced tumor suppressor genes in patients with myelodysplastic syndromes (MDS). It is unclear whether azacytidine can alter the proliferative and apoptotic changes in myeloid leukemia cells, and methylation changes induced by this drug have remained poorly characterized in therapy-related models. METHODS: The proliferation rate of azacytidine on HL60 cells was determined by the MTT protocol. Methylation-specific PCR (MSP) and RT-PCR were used respectively to detect gene methylation status changes and expression levels of p16, Death associated protein kinase (DAPK) and O(6)-methylguanine-DNA methyltransferase (MGMT) before and after treatment with azacytidine. RESULTS: Azacytidine inhibited HL60 cell proliferation and showed a time- and dose-dependent effect. MSP showed hypermethylated p16, DAPK, and MGMT genes before azacytidine treatment. Complete demethylation was seen in p16 and DAPK genes and partial demethylation in the MGMT gene after co-culture with azacytidine. The expression level of p16, DAPK and MGMT genes in HL60 cells was up-regulated after treatment with azacytidine. CONCLUSIONS: The CpG islands of p16, DAPK and MGMT genes are hypermethylated in HL60 cells. Azacytidine inhibits proliferation of leukemic cells by hypomethylation of p16, DAPK and MGMT genes.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Azacitidina/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Genes p16/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Islas de CpG/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular , Genes Reguladores/efectos de los fármacos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/metabolismo
15.
Cancer Epidemiol Biomarkers Prev ; 21(1): 182-90, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22028397

RESUMEN

BACKGROUND: Sufficient epidemiologic evidence shows an etiologic link between polycyclic aromatic hydrocarbons (PAH) exposure and lung cancer risk. While the genetic modifications have been found in PAH-exposed population, it is unclear whether gene-specific methylation involves in the process of PAH-associated biologic consequence. METHODS: Sixty-nine PAH-exposed workers and 59 control subjects were recruited. Using bisulfite sequencing, we examined the methylation status of p16(INK4α) promoter in peripheral blood lymphocytes (PBL) from PAH-exposed workers and in benzo(a)pyrene (BaP)-transformed human bronchial epithelial (HBE) cells. The relationships between p16(INK4α) methylation and the level of urinary 1-hydroxypyrene (1-OHP) or the frequency of cytokinesis block micronucleus (CBMN) were analyzed. RESULTS: Compared with the control group, PAH-exposed workers exhibited higher levels of urinary 1-OHP (10.62 vs. 2.52 µg/L), p16(INK4α) methylation (7.95% vs. 1.14% for 22 "hot" CpG sites), and CBMN (7.28% vs. 2.92%) in PBLs. p16(INK4α) hypermethylation in PAH-exposed workers exhibited CpG site specificity. Among the 35 CpG sites we analyzed, 22 were significantly hypermethylated. These 22 hypermethylated CpG sites were positively correlated to levels of urinary 1-OHP and CBMN in PBLs. Moreover, the hypermethylation and suppression of p16 expression was also found in BaP-transformed HBER cells. CONCLUSION: PAH exposure induced CpG site-specific hypermethylation of p16(INK4α) gene. The degree of p16(INK4α) methylation was associated with the levels of DNA damage and internal exposure. IMPACT: p16(INK4α) hypermethylation might be an essential biomarker for the exposure to PAHs and for early diagnosis of cancer.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Metilación de ADN , Genes p16/efectos de los fármacos , Linfocitos/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/sangre , Hidrocarburos Policíclicos Aromáticos/envenenamiento , Adulto , Biomarcadores/sangre , Biomarcadores/metabolismo , Biomarcadores/orina , Línea Celular , Islas de CpG , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Linfocitos/fisiología , Pruebas de Micronúcleos/métodos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Exposición Profesional/efectos adversos , Exposición Profesional/análisis , Regiones Promotoras Genéticas , Pirenos/metabolismo , Factores de Riesgo
16.
Am J Hypertens ; 25(3): 354-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22113172

RESUMEN

BACKGROUND: We recently reported that aldosterone-induced cellular senescence via an increase in p21, a cyclin-dependent kinase (CDK) inhibitor, in rat kidney and cultured human proximal tubular cells. In the present study, we investigated the contribution of aldosterone to the renal p21 expression and senescence during the development of angiotensin II (AngII)-induced hypertension. METHODS: Mice received 1% salt in drinking water and vehicle or AngII, and were divided into five groups: 1, vehicle; 2, AngII; 3, AngII+olmesartan; 4, AngII+eplerenone; and 5, AngII+hydralazine. RESULTS: Plasma aldosterone levels were increased by AngII infusion. Eplerenone further elevated the plasma aldosterone level, but olmesartan and hydralazine did not. AngII group showed significant increase in blood pressure compared to vehicle. Olmesartan and hydralazine, but not eplerenone, suppressed the AngII-salt hypertension. The increase in urinary protein excretion by AngII-salt was suppressed only by olmesartan. AngII with high salt induced a greater expression of p21 mRNA in the kidney than vehicle. Olmesartan abolished the increase in p21 expression, whereas neither eplerenone nor hydralazine affected it. AngII with high salt did not change the expression of p16, another CDK inhibitor. The mice lacking p21 showed identical changes on blood pressure and albuminuria in response to AngII with high salt compared to wild type. CONCLUSION: These results suggest that aldosterone does not predominantly contribute to renal p21 expression and senescence during the development of AngII-salt hypertension, and that the increase in p21 in the kidney is not likely involved in the development of hypertension and albuminuria.


Asunto(s)
Aldosterona/fisiología , Angiotensina II/farmacología , Presión Sanguínea/efectos de los fármacos , Senescencia Celular/genética , Hipertensión/genética , Riñón/metabolismo , Quinasas p21 Activadas/genética , Actinas/efectos de los fármacos , Animales , Antihipertensivos/farmacología , Eplerenona , Genes p16/efectos de los fármacos , Genes p53/efectos de los fármacos , Hidralazina/farmacología , Hipertensión/inducido químicamente , Imidazoles/farmacología , Ratones , Ratones Endogámicos C57BL , Sirtuina 1/efectos de los fármacos , Sirtuina 1/genética , Cloruro de Sodio/farmacología , Espironolactona/análogos & derivados , Espironolactona/farmacología , Tetrazoles/farmacología
17.
Braz J Med Biol Res ; 44(11): 1118-24, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21971687

RESUMEN

The testicular feminized (Tfm) mouse carries a nonfunctional androgen receptor (AR) and reduced circulating testosterone levels. We used Tfm and castrated mice to determine whether testosterone modulates markers of aging in cardiomyocytes via its classic AR-dependent pathway or conversion to estradiol. Male littermates and Tfm mice were divided into 6 experimental groups. Castrated littermates (group 1) and sham-operated Tfm mice (group 2, N = 8 each) received testosterone. Sham-operated Tfm mice received testosterone in combination with the aromatase inhibitor anastrazole (group 3, N = 7). Castrated littermates (group 4) and sham-operated untreated Tfm mice (group 5) were used as controls (N = 8 and 7, respectively). An additional control group (group 6) consisted of age-matched non-castrated littermates (N = 8). Cardiomyocytes were isolated from the left ventricle, telomere length was measured by quantitative PCR and expression of p16INK4α, retinoblastoma (Rb) and p53 proteins was detected by Western blot 3 months after treatment. Compared with group 6, telomere length was short (P < 0.01) and expression of p16INK4α, Rb and p53 proteins was significantly (P < 0.05) up-regulated in groups 4 and 5. These changes were improved to nearly normal levels in groups 1 and 2 (telomere length = 0.78 ± 0.05 and 0.80 ± 0.08; p16INK4α = 0.13 ± 0.03 and 0.15 ± 0.04; Rb = 0.45 ± 0.05 and 0.39 ± 0.06; p53 = 0.16 ± 0.04 and 0.13 ± 0.03), but did not differ between these two groups. These improvements were partly inhibited in group 3 compared with group 2 (telomere length = 0.65 ± 0.08 vs 0.80 ± 0.08, P = 0.021; p16INK4α = 0.28 ± 0.05 vs 0.15 ± 0.04, P = 0.047; Rb = 0.60 ± 0.06 vs 0.39 ± 0.06, P < 0.01; p53 = 0.34 ± 0.06 vs 0.13 ± 0.03, P = 0.004). In conclusion, testosterone deficiency contributes to cardiomyocyte aging. Physiological testosterone can delay cardiomyocyte aging via an AR-independent pathway and in part by conversion to estradiol.


Asunto(s)
Envejecimiento/metabolismo , Senescencia Celular/fisiología , Estradiol/metabolismo , Miocitos Cardíacos/fisiología , Receptores Androgénicos/metabolismo , Testosterona/farmacología , Envejecimiento/patología , Animales , Biomarcadores/análisis , Genes p16/efectos de los fármacos , Masculino , Ratones , Modelos Animales , Orquiectomía , Distribución Aleatoria , Proteína de Retinoblastoma/metabolismo , Acortamiento del Telómero/efectos de los fármacos , Testosterona/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
18.
Epigenetics ; 6(9): 1114-9, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21822058

RESUMEN

Changes in DNA methylation patterns are a hallmark of tobacco-induced carcinogenesis. We have conducted a randomized 4-week intervention trial to investigate the effects of three dietary regimens to modify DNA methylation patterns in peripheral white blood cells of heavy smokers. A group of 88 smokers were randomly assigned to and distributed among three diets, including (1) normal isocaloric diet (balanced in fruits and vegetables), according to international guidelines; (2) a diet enriched in flavonoids and isothiocyanates (particularly cruciferous vegetables); (3) a regimen consisting of diet 1 supplemented with flavonoids (green tea and soy products). Methylation patterns were analyzed by pyrosequencing in LINE1 (Long Interspersed DNA Elements), RASSF1A, ARF and CDKN2a (tumor suppressor genes), MLH1 (mismatch DNA repair) and MTHFR (folate metabolism). Three distinct patterns of methylation were observed. In LINE1, methylation showed a small but reproducible increase with all three regimens. MTHFR was constitutively methylated with no significant modulation by diets. The four other loci showed low basal levels of methylation with no substantial change after intervention. These data suggest that the isocaloric diet may stabilize global epigenetic (LINE1 DNA methylation) patterns in peripheral white blood cells but does not provide evidence for methylation changes in specific genes associated with this short-term dietary intervention.


Asunto(s)
Brassicaceae/química , Metilación de ADN , Dieta , Leucocitos/efectos de los fármacos , Fumar/sangre , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Epigénesis Genética , Flavonoides/administración & dosificación , Flavonoides/farmacología , Genes p16/efectos de los fármacos , Humanos , Isotiocianatos/administración & dosificación , Isotiocianatos/farmacología , Leucocitos/química , Leucocitos/citología , Elementos de Nucleótido Esparcido Largo , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/química , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/química , Proteínas Nucleares/genética , Análisis de Secuencia de ADN/métodos , Fumar/genética , Glycine max/química , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
19.
J Dent Res ; 90(1): 65-70, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20940365

RESUMEN

Several studies have reported Oral Squamous Cell Carcinoma (OSCC) association with etiological factors, such as smoking and alcohol. The aim of the present study was to establish whether the methylenetetrahydrofolate reductase (MTHFR) C677T genotype and a high alcohol intake, solely or in interaction, have an impact on the oral cancer risk, DNA methylation, or multiple methylation of tumor-related genes. MTHFR C677T genetic polymorphism was determined by the PCR/RFLP method, and DNA methylation was assessed by nested methylation-specific PCR. The risk for multiple methylation was significantly increased in heavy-drinking patients with the TT genotype, compared with CC and CT patients (OR = 10.873; 95% CI, 1.134-104.24). Multiple methylation was significantly associated with tumor stage (p = 0.018), and showed a trend of association with the presence of nodal metastases (p = 0.058). A significant association was found between TT genotype and methylation status of the RASSF1A gene in OSCC patients (p = 0.012). Heavy-drinking individuals with the TT genotype showed increased oral cancer risk compared with the CC genotype (OR = 3.601; 95% CI, 1.036-12.513), and compared with the CC and CT genotypes (OR = 4.288; 95% CI, 1.325-13.877). Our study suggested gene-environment interactions between high alcohol intake and the MTHFR 677TT genotype for elevated oral cancer risk, with a significant impact on multiple methylation of cancer-related genes.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Carcinoma de Células Escamosas/etiología , Citosina , Metilación de ADN/efectos de los fármacos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Neoplasias de la Boca/etiología , Oncogenes/efectos de los fármacos , Polimorfismo Genético/genética , Timina , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Anciano de 80 o más Años , Cadherinas/efectos de los fármacos , Cadherinas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundario , Estudios de Casos y Controles , Estudios de Cohortes , Metilación de ADN/genética , Femenino , Genes p16/efectos de los fármacos , Genotipo , Humanos , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Estadificación de Neoplasias , Oncogenes/genética , Proteínas Represoras/efectos de los fármacos , Proteínas Represoras/genética , Factores de Riesgo , Factores Sexuales , Fumar/efectos adversos , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/genética
20.
Anticancer Res ; 30(12): 4907-13, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21187469

RESUMEN

Pancreatic cancer has a poor prognosis, mainly due to lack of effective therapies. This study demonstrated the ability of dietary agent, indole-3-carbinol (I3C), to lower the LD(50) of gemcitabine (Gemzar) in decreasing growth of both male (MiaPaca2) and female (SU86.86) pancreatic cancer cells. Female pancreatic cancer cells were more resistant to gemcitabine alone. Additionally, RT-PCR analysis of MiaPaca2 cells treated with 1, 10 or 100 µM of I3C showed that I3C reactivated the tumor suppressor gene p16INK4a in pancreatic cancer cells. Methylated-specific PCR analysis indicated that I3C demethylated the promoter region of p16 INK4a, which was methylated in the untreated cancer cells. p16INK4a inactivation through promoter hypermethylation is considered an early event in pancreatic carcinogenesis. A positive control using 5-azacytidine also reactivated p16INK4a. This study demonstrated the potential of I3C, a possible non-toxic hypomethylating agent, combined with the anticancer agent, gemcitabine, to be a powerful strategy for treating pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Indoles/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Línea Celular Tumoral , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes p16/efectos de los fármacos , Humanos , Indoles/administración & dosificación , Masculino , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA