Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chem Res Toxicol ; 33(2): 625-633, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31841318

RESUMEN

DNA methylating agents are abundant in the environment and are sometimes used in cancer chemotherapy. They react with DNA to form methyl-DNA adducts and byproduct lesions that can be both toxic and mutagenic. Foremost among the mutagenic lesions is O6-methylguanine (m6G), which base pairs with thymine during replication to cause GC → AT mutations. The gpt delta C57BL/6J mouse strain of Nohmi et al. (Mol. Mutagen 1996, 28, 465-70) reliably produces mutational spectra of many DNA damaging agents. In this work, mouse embryo fibroblasts (MEFs) were made from gpt delta C57BL/6J mice and evaluated as a screening tool to determine the qualitative and quantitative features of mutagenesis by N-methyl-N-nitrosourea (MNU), a direct-acting DNA alkylator that serves as a model for environmental N-nitrosamines, such as N-nitrosodimethylamine and therapeutic agents such as Temozolomide. The DNA repair protein MGMT (O6-methylguanine DNA methyltransferase) protects against environmental mutagenesis by DNA methylating agents and, by removing m6G, limits the therapeutic potential of Temozolomide in cancer therapy. The gpt delta MEFs were treated with MNU to establish dose-dependent toxicity. In parallel, MNU mutagenicity was determined in the presence and absence of the MGMT inhibitor AA-CW236 (4-(2-(5-(chloromethyl)-4-(4-(trifluoromethoxy)phenyl)-1H-1,2,3-triazol-1-yl)ethyl)-3,5-dimethylisoxazole). With and without the inhibitor, the principal mutagenic event of MNU was GC → AT, but more mutations were observed when the inhibitor was present. Evidence that the mutagenic lesion was m6G was based on mass spectral data collected using O6-methyl-d3-guanine as an internal standard; m6G levels were higher in AA-CW236 treated MEFs by an amount proportional to the higher mutation frequency seen in the same cells. This work establishes gpt delta MEFs as a versatile tool for probing mutagenesis by environmental and therapeutic agents and as a cell culture model in which chemical genetics can be used to determine the impact of DNA repair on biological responses to DNA damaging agents.


Asunto(s)
Alquilantes/farmacología , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Metilnitrosourea/farmacología , Mutagénesis/efectos de los fármacos , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Alquilantes/química , Animales , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Inhibidores Enzimáticos/química , Fibroblastos/metabolismo , Metilnitrosourea/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Supresoras de Tumor/metabolismo
2.
Toxicol Rep ; 6: 426-430, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31193040

RESUMEN

Andrographis paniculata is widely used in traditional herbal medicines for the treatment of common cold, fever and diarrhea, in many regions of Scandinavia and Asia, including Thailand. The pharmacological activities of A. paniculata are mainly attributed to active diterpenoids including 14-deoxyandrographolide, which is uniquely high in first true leaf ethanolic extract (FTLEE) of A. paniculata. In this study, the acute toxicity of the standardized FTLEE of A. paniculata was examined according to the OECD test guideline No. 420. Mice were divided into four groups of each sex and orally received the standardized FTLEE of A. paniculata (0, 300, 2000, or 5000 mg/kg BW). Post-treatment, body weight, signs of toxicity, and/or mortality were observed for 14 days. At Day 15, animals were euthanized, internal organs were observed grossly, and blood samples collected were subjected to hematology and clinical biochemistry analyses. The results showed that all treated animals survived and no apparent adverse effects were observed during the duration of the study. Gross necropsy observation revealed no lesion in any organ of all the standardized FTLEE-treated mice. Although significant alterations in BUN, lymphocytes, neutrophils, hematocrit and hemoglobin were observed, these alterations were not treatment-related toxic effects. Therefore, we concluded that a single oral administration of the standardized FTLEE of A. paniculata with an upper fixed dose of 5000 mg/kg BW has no significant acute toxicological effects.

3.
Food Chem Toxicol ; 118: 595-607, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29890199

RESUMEN

Previous studies showed that glyphosate stimulates breast cancer cell growth via estrogen receptors. The present study investigated the effect of glyphosate on the estrogen signaling pathway involved in the induction of cholangiocarcinoma (CCA) cell growth. HuCCA-1, RMCCA-1 and MMNK-1 were chosen for comparison. The effects of glyphosate on cell growth, cell cycle and molecular signaling pathways were measured. The results showed that HuCCA-1 cells expressed estrogen receptor alpha (ERα), while ERα was not detected in RMCCA-1 and MMNK-1 cells. ERα was mostly expressed in cytoplasmic compartment of HuCCA-1 cells. Estradiol (E2) (10-11-10-5 M) induced cell proliferation in HuCCA-1 but not in RMCCA-1 and MMNK-1 cells. Glyphosate at the same concentration range also induced HuCCA-1 cell proliferation. The S phase of the cell cycle, and protein levels of the cyclin family were significantly increased after treatment of glyphosate or E2. Both compounds also induced the expression of proliferative signaling-related proteins including ERα, VEGFR2, pERK, PI3K(p85), and PCNA. These effects of glyphosate and E2 were abolished by the ER antagonist, 4-hydroxytamoxifen and U0126, a MEK inhibitor. The data from this study indicate that glyphosate can induce cell growth in ERα positive CCA cells through non-genomic estrogen receptor/ERK1/2 signaling pathway.


Asunto(s)
Colangiocarcinoma/patología , Receptor alfa de Estrógeno/efectos de los fármacos , Glicina/análogos & derivados , Herbicidas/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Colangiocarcinoma/metabolismo , Receptor alfa de Estrógeno/metabolismo , Glicina/toxicidad , Humanos , Transducción de Señal/efectos de los fármacos , Glifosato
4.
Int J Oncol ; 53(1): 177-188, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29693152

RESUMEN

Cholangiocarcinoma (CCA) is a malignant tumor of the biliary epithelium associated with Opisthorchis viverrini, primary sclerosing cholangitis and hepatitis viral infection. In the global population, men have higher incidence rates for CCA than women; thus, a gender disparity in the progression of chronic inflammation of the biliary duct leading to malignancy may involve the effects of estrogen (E2). Genistein (GE), a prominent phytoestrogen found in soy products, is an estrogen receptor ß (ERß) agonist and a tyrosine kinase inhibitor. The present study investigated the effects of GE on the growth of CCA cells by cell viability assay. The effects on signaling proteins were detected by western blot analysis and ELISA. Gene expression was examined by RT-qPCR. Two human intrahepatic CCA cell lines, HuCCA­1 and RMCCA­1, were utilized. GE (50­200 µM) reduced the viability of the two cell lines, and also inhibited the activation of epidermal growth factor receptor (EGFR) and AKT, as evidenced by decreasing protein levels of phosphorylated (p)-EGFR (Tyr1173) and p­AKT (Ser473), respectively. GE altered the mitogen­activated protein kinase signaling cascade by mediating decreased protein levels of p­p38 and increased protein levels of p­ERK1/2. GE significantly decreased the levels of interleukin 6 (IL6) and induced the expression of inducible nitric oxide synthase (iNOS). GE also downregulated the expression of p­ERα (Ser118) protein and ERα mRNA levels. Finally, GE induced the downregulation of the protein levels of ERß. Of note, E2 deprivation potentiated the GE-induced reduction of p­EGFR (Tyr1173) and total AKT proteins and production of IL6, and mediated the downregulation of GE-induced iNOS protein. In conclusion, GE inhibited the growth of human CCA cell lines by reducing the activation of EGFR and AKT, and by attenuating the production of IL6. E2 and ER were also involved in the growth-inhibitory effect of GE in CCA cells.


Asunto(s)
Colangiocarcinoma/tratamiento farmacológico , Receptores ErbB/genética , Genisteína/administración & dosificación , Interleucina-6/genética , Proteína Oncogénica v-akt/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Receptores ErbB/antagonistas & inhibidores , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Oncogénica v-akt/antagonistas & inhibidores , Fosforilación , Transducción de Señal/efectos de los fármacos
5.
Planta Med ; 83(9): 778-789, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28249303

RESUMEN

Andrographis paniculata has been widely used in Scandinavian and Asian counties for the treatment of the common cold, fever, and noninfectious diarrhea. The present study was carried out to investigate the physiological effects of short-term multiple dose administration of a standardized A. paniculata capsule used for treatment of the common cold and uncomplicated upper respiratory tract infections, including blood pressure, electrocardiogram, blood chemistry, hematological profiles, urinalysis, and blood coagulation in healthy Thai subjects. Twenty healthy subjects (10 males and 10 females) received 12 capsules per day orally of 4.2 g of a standardized A. paniculata crude powder (4 capsules of 1.4 g of A. paniculata, 3 times per day, 8 h intervals) for 3 consecutive days. The results showed that all of the measured clinical parameters were found to be within normal ranges for a healthy person. However, modulation of some parameters was observed after the third day of treatment, for example, inductions of white blood cells and absolute neutrophil count in the blood, a reduction of plasma alkaline phosphatase, and an induction of urine pH. A rapid and transient reduction in blood pressure was observed at 30 min after capsule administration, resulting in a significant reduction of mean systolic blood pressure. There were no serious adverse events observed in the subjects during the treatment period. In conclusion, this study suggests that multiple oral dosing of A. paniculata at the normal therapeutic dose for the common cold and uncomplicated upper respiratory tract infections modulates various clinical parameters within normal ranges for a healthy person.


Asunto(s)
Andrographis , Coagulación Sanguínea/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Preparaciones de Plantas/farmacología , Administración Oral , Adulto , Análisis Químico de la Sangre , Cápsulas , Femenino , Humanos , Masculino , Fitoterapia , Preparaciones de Plantas/administración & dosificación , Pulso Arterial , Tailandia
6.
J Ethnopharmacol ; 179: 156-61, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26721218

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Flower, seed, and fruit of longan (Dimocarpus longan Lour.) have been used in the Traditional Chinese Medicine (TCM) serving as a common herb in relief of swelling which can be applied in cases of inflammatory diseases. However, the scientific evidence related to their effects on inflammation especially the possible cellular and molecular mechanisms of longan need to be clarified. AIM OF THE STUDY: To evaluate the anti-inflammatory effect of the various parts of longan including flower, seed, and pulp. The mechanisms and molecular targets involved in their effects were also investigated. MATERIALS AND METHODS: Different longan extracts were analyzed for their bioactive compounds and evaluated for anti-inflammation. Corilagin, ellagic acid, and gallic acid were detected using HPLC-DAD. In vitro anti-inflammatory effect of longan extracts and their polysaccharides were examined by analyzing nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Expression of the proteins that involved in NO production was detected by Western blot. RESULTS: Flower extract contained the highest amounts of total phenolics, total flavonoids, proanthocyanidins, corilagin and ellagic acid when compared to seed and pulp extracts. The extracts of different longan parts inhibited LPS-induced NO production with different potency. The highest potency for the inhibition of NO production was shown with flower extract follow by seed and pulp (IC50=128.2, 1127.4, and 1260.2µgmL(-1), respectively). The mechanisms of this anti-NO production were associated with their NO scavenging effect and their decreasing the expression and catalytic activity of an inducible nitric oxide synthase (iNOS). Moreover, these longan extracts suppressed LPS-induced degradation of IκBα and activation of NF-κB, activator protein-1 (AP-1), Akt, and mitogen activated protein kinases (MAPKs). CONCLUSION: These results suggest that the longan extracts possess anti-inflammatory property. Therefore, longan could provide potential dietary supplement for the treatment of inflammatory-related diseases.


Asunto(s)
Lipopolisacáridos/antagonistas & inhibidores , Macrófagos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , Sapindaceae/química , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Animales , Flores/química , Frutas/química , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Ratones , FN-kappa B/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Extractos Vegetales/farmacología , Polisacáridos/química , Polisacáridos/farmacología , Células RAW 264.7 , Semillas/química , Factor de Transcripción AP-1/efectos de los fármacos
7.
Environ Health Perspect ; 124(2): 170-5, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26115410

RESUMEN

BACKGROUND: Millions of individuals worldwide, particularly those living in rural and developing areas, are exposed to harmful levels of inorganic arsenic (iAs) in their drinking water. Inorganic As exposure during key developmental periods is associated with a variety of adverse health effects, including those that are evident in adulthood. There is considerable interest in identifying the molecular mechanisms that relate early-life iAs exposure to the development of these latent diseases, particularly in relationship to cancer. OBJECTIVES: This work summarizes research on the molecular mechanisms that underlie the increased risk of cancer development in adulthood that is associated with early-life iAs exposure. DISCUSSION: Epigenetic reprogramming that imparts functional changes in gene expression, the development of cancer stem cells, and immunomodulation are plausible underlying mechanisms by which early-life iAs exposure elicits latent carcinogenic effects. CONCLUSIONS: Evidence is mounting that relates early-life iAs exposure and cancer development later in life. Future research should include animal studies that address mechanistic hypotheses and studies of human populations that integrate early-life exposure, molecular alterations, and latent disease outcomes.


Asunto(s)
Arsénico/toxicidad , Exposición a Riesgos Ambientales , Neoplasias/inducido químicamente , Contaminantes Químicos del Agua/toxicidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Agua Potable/análisis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Factores de Riesgo , Adulto Joven
8.
J Appl Toxicol ; 36(6): 790-801, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26234195

RESUMEN

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are the two most popular surfactants among perfluorinated compounds (PFCs), with a wide range of uses. Growing evidence suggests that PFCs have the potential to interfere with estrogen homeostasis, posing a risk of endocrine-disrupting effects. This in vitro study aimed to investigate the estrogenic effect of these compounds on T47D hormone-dependent breast cancer cells. PFOS and PFOA (10(-12) to 10(-4) M) were not able to induce estrogen response element (ERE) activation in the ERE luciferase reporter assay. The ERE activation was induced when the cells were co-incubated with PFOS (10(-10) to 10(-7) M) or PFOA (10(-9) to 10(-7) M) and 1 nM of 17ß-estradiol (E2). PFOS and PFOA did not modulate the expression of estrogen-responsive genes, including progesterone (PR) and trefoil factor (pS2), but these compounds enhanced the effect of E2-induced pS2 gene expression. Neither PFOS nor PFOA affected T47D cell viability at any of the tested concentrations. In contrast, co-exposure with PFOS or PFOA and E2 resulted in an increase of E2-induced cell viability, but no effect was found with 10 ng ml(-1) EGF co-exposure. Both compounds also intensified E2-dependent growth in the proliferation assay. ERK1/2 phosphorylation was increased by co-exposure with PFOS or PFOA and E2, but not with EGF. Collectively, this study shows that PFOS and PFOA did not possess estrogenic activity, but they enhanced the effects of E2 on estrogen-responsive gene expression, ERK1/2 activation and the growth of the hormone-deprived T47D cells. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Neoplasias de la Mama/inducido químicamente , Caprilatos/toxicidad , Disruptores Endocrinos/toxicidad , Estradiol/agonistas , Estrógenos/agonistas , Fluorocarburos/toxicidad , Tensoactivos/toxicidad , Ácidos Alcanesulfónicos/antagonistas & inhibidores , Butadienos/farmacología , Caprilatos/antagonistas & inhibidores , Carcinógenos Ambientales/química , Carcinógenos Ambientales/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disruptores Endocrinos/química , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Fluorocarburos/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nitrilos/farmacología , Concentración Osmolar , Inhibidores de Proteínas Quinasas/farmacología , Elementos de Respuesta/efectos de los fármacos , Tensoactivos/química , Factor Trefoil-1/agonistas , Factor Trefoil-1/genética , Factor Trefoil-1/metabolismo
9.
Environ Toxicol ; 31(9): 1133-46, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25728338

RESUMEN

Arsenic (As) is considered a major environmental health threat worldwide due to its widespread contamination in drinking water. Recent studies reported that arsenic is a potential xenoestrogen as it interfered with the action of estrogen (E2) and estrogen receptor (ER) signaling. The present study investigated the effects of sodium arsenite (NaAsO2 ) on estrogen signaling in human breast cancer cells. The results demonstrated that NaAsO2 dose-dependently increased viability of hormone-dependent breast cancer MCF-7 and T47D cells expressing both ERα and ERß but not hormone-independent MDA-MB-231 cells expressing ERß. These suggested ERα contribution to NaAsO2 -stimulated breast cancer cells growth. NaAsO2 induced down-regulation of ERα but up-regulation of ERß protein expressions in T47D cells. Moreover, NaAsO2 dose-dependently inhibited E2-induced ER transcriptional activity as it decreased E2-mediated ERE-luciferase transcription activation and PgR mRNA transcription but increased pS2 mRNA transcription. However, NaAsO2 induced both rapid and sustained activation of ERK1/2 and increased in phosphorylation of ERα at serine 118 residue, c-fos and c-myc protein expressions. These results indicated that NaAsO2 interferes the genomic estrogen-signaling pathway but induces activation of a rapid nongenomic signal transduction through ERK1/2 pathway which may contribute to its proliferative effect on hormone-dependent breast cancer cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1133-1146, 2016.


Asunto(s)
Arsenitos/toxicidad , Receptor alfa de Estrógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Compuestos de Sodio/toxicidad , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Células MCF-7 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Presenilina-2/genética , Presenilina-2/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
Toxicol Lett ; 236(3): 138-44, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25982963

RESUMEN

Arsenic neurotoxicity has a broad range of adverse effects on human health, which are induced in part by inhibition of neurite outgrowth. Insulin has been reported to promote neurite extension. The present study investigated whether insulin can protect neurons from impaired neurite outgrowth induced by arsenic, and examined the signaling pathway involved in this action. The study demonstrated that NaAsO2 caused inhibition of neurite outgrowth in differentiated SH-SY5Y cells indicating its neurotoxicity. This inhibitory effect of NaAsO2 was attenuated by insulin. It was found that blocking PI3K or Akt by selective inhibitors canceled the protective effect of insulin against NaAsO2-induced neurite outgrowth impairment suggesting the essential role of active PI3K and Akt in insulin's protective action. Inhibition of GSK3, which mimics an effect of insulin stimulation, had no effect on the impairment of neurite outgrowth by NaAsO2 implying that the insulin protective action is probably not due to its mediation of GSK3 inhibition ability. Moreover, NaAsO2 decreased the Akt activity, as it caused reduction in Akt phosphorylation, and downregulated expression of SIRT1. Additionally, the reduction of these signals by NaAsO2 was attenuated by insulin. Taken together, these results show that insulin attenuates arsenic-induced neurite outgrowth impairment possibly via activation of PI3K/Akt/SIRT1 signaling, and arsenic may exert neurite outgrowth inhibition through a mechanism involving reduction of signaling molecules downstream from insulin, PI3K/Akt/SIRT1. Our findings raise the possibility of using insulin to combat arsenic neurotoxicity.


Asunto(s)
Arsenitos/toxicidad , Insulina/farmacología , Neuritas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirtuina 1/metabolismo , Compuestos de Sodio/toxicidad , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neuritas/fisiología , Neuroblastoma , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/genética , Sirtuina 1/genética
11.
Int J Oncol ; 46(6): 2317-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25815516

RESUMEN

Cholangiocarcinoma (CCA) is a malignant cancer of the biliary tract and its occurrence is associated with chronic cholestasis which causes an elevation of bile acids in the liver and bile duct. The present study aimed to investigate the role and mechanistic effect of bile acids on the CCA cell growth. Intrahepatic CCA cell lines, RMCCA-1 and HuCCA-1, were treated with bile acids and their metabolites to determine the growth promoting effect. Cell viability, cell cycle analysis, EdU incorporation assays were conducted. Intracellular signaling proteins were detected by western immunoblotting. Among eleven forms of bile acids and their metabolites, only taurolithocholic acid (TLCA) concentration dependently (1-40 µM) increased the cell viability of RMCCA-1, but not HuCCA-1 cells. The cell cycle analysis showed induction of cells in the S phase and the EdU incorporation assay revealed induction of DNA synthesis in the TLCA-treated RMCCA-1 cells. Moreover, TLCA increased the phosphorylation of EGFR, ERK 1/2 and also increased the expression of cyclin D1 in RMCCA-1 cells. Furthermore, TLCA-induced RMCCA-1 cell growth could be inhibited by atropine, a non-selective muscarinic acetylcholine receptor (mAChR) antagonist, AG 1478, a specific EGFR inhibitor, or U 0126, a specific MEK 1/2 inhibitor. These results suggest that TLCA induces CCA cell growth via mAChR and EGFR/EKR1/2 signaling pathway. Moreover, the functional presence of cholinergic system plays a certain role in TLCA-induced CCA cell growth.


Asunto(s)
Ácidos y Sales Biliares/efectos adversos , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ácido Taurolitocólico/efectos adversos , Ácidos y Sales Biliares/farmacología , Neoplasias de los Conductos Biliares/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Colangiocarcinoma/metabolismo , Ciclina D1/metabolismo , Receptores ErbB/metabolismo , Humanos , Fosforilación , Receptores Muscarínicos , Ácido Taurolitocólico/farmacología
12.
Int J Cancer ; 136(6): 1254-62, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25070670

RESUMEN

It has become axiomatic that critical windows of susceptibility to genotoxins exist and that genetic damage in utero may be a trigger for later life cancers. Data supporting this critical window hypothesis are remarkably few. This study provides a quantitative bridge between DNA damage by the liver carcinogen aflatoxin B1 (AFB1 ) during prenatal development and the risk of later life genetic disease. AFB1 was given to pregnant C57BL/6J mice, carrying F1 gestation day 14 (GD14) embryos of the B6C3F1 genotype. Ultra-high performance liquid chromatography and mass spectrometry (UPLC-MS) using aflatoxin-(15) N5 -guanine adduct standards afforded measurement of the AFB1 -N(7) -Gua and AFB1 -FAPY adducts 6-hr post dosing in liver DNA of mothers and embryos. A parallel cohort gave birth and the livers of the F1 were analyzed for mutations in the gpt gene at 3 and 10 weeks of age. The data revealed mutational spectra dominated by G:C to T:A mutations in both the mother and offspring that are characteristic of AFB1 and distinct from background. It was shown that adducts in GD14 embryos were 20-fold more potent inducers of mutagenesis than adducts in parallel-dosed adults. This sensitivity enhancement correlated with Ki67 staining of the liver, reflecting the proliferative potential of the tissue. Taken together, these data provide insight into the relative genetic risks of prenatal and adult exposures to AFB1 . Early life exposure, especially during the embryonic period, is strikingly more mutagenic than treatment later in life. Moreover the data provide a baseline against which risk prevention strategies can be evaluated.


Asunto(s)
Aflatoxina B1/toxicidad , Carcinógenos/toxicidad , Feto/efectos de los fármacos , Hígado/efectos de los fármacos , Mutación , Animales , Proliferación Celular/efectos de los fármacos , Aductos de ADN/análisis , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL
13.
Planta Med ; 80(7): 533-43, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24782229

RESUMEN

Andrographis paniculata is an important herbal medicine widely used in several Asian countries for the treatment of various diseases due to its broad range of pharmacological activities. The present study reports that A. paniculata extracts potently inhibit the growth of liver (HepG2 and SK-Hep1) and bile duct (HuCCA-1 and RMCCA-1) cancer cells. A. paniculata extracts with different contents of major diterpenoids, including andrographolide, 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, and 14-deoxyandrographolide, exhibited a different potency of growth inhibition. The ethanolic extract of A. paniculata at the first true leaf stage, which contained a high amount of 14-deoxyandrographolide but a low amount of andrographolide, showed a cytotoxic effect to cancer cells about 4 times higher than the water extract of A. paniculata at the mature leaf stage, which contained a high amount of andrographolide but a low amount of 14-deoxyandrographolide. Andrographolide, not 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, or 14-deoxyandrographolide, possessed potent cytotoxic activity against the growth of liver and bile duct cancer cells. The cytotoxic effect of the water extract of A. paniculata at the mature leaf stage could be explained by the present amount of andrographolide, while the cytotoxic effect of the ethanolic extract of A. paniculata at the first true leaf stage could not. HuCCA-1 cells showed more sensitivity to A. paniculata extracts and andrographolide than RMCCA-1 cells. Furthermore, the ethanolic extract of A. paniculata at the first true leaf stage increased cell cycle arrest at the G0/G1 and G2/M phases, and induced apoptosis in both HuCCA-1 and RMCCA-1 cells. The expressions of cyclin-D1, Bcl-2, and the inactive proenzyme form of caspase-3 were reduced by the ethanolic extract of A. paniculata in the first true leaf stage treatment, while a proapoptotic protein Bax was increased. The cleavage of poly (ADP-ribose) polymerase was also found in the ethanolic extract of A. paniculata in the first true leaf stage treatment. This study suggests that A. paniculata could be a promising herbal plant for the alternative treatment of intrahepatic cholangiocarcinoma.


Asunto(s)
Andrographis/química , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Diterpenos/uso terapéutico , Fitoterapia , Extractos Vegetales/uso terapéutico , Apoptosis/efectos de los fármacos , Conductos Biliares Intrahepáticos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diterpenos/química , Diterpenos/aislamiento & purificación , Humanos , Concentración 50 Inhibidora , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales
14.
Environ Toxicol ; 29(6): 672-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22859221

RESUMEN

Arsenic is a widespread contaminant in the environment especially in drinking water. Although it is a known carcinogen in human, the mechanism by which arsenic induces carcinogenesis is not well understood. Among several effects of arsenic, it has been suggested that arsenic-induced vascular endothelial growth factor (VEGF) expression plays a critical role in arsenic carcinogenesis. In the present study, we demonstrated that arsenite induced VEGF expression in neuroblastoma SH-SY5Y cells without induction of HIF-1α, a well-known transcriptional activator for VEGF suggesting that arsenite-induced VEGF expression in SH-SY5Y cells may not require HIF-1α activation. It has been reported that VEGF expression is regulated by multiple transcription factors including ß-catenin. We therefore investigated whether ß-catenin was involved in arsenite-induced VEGF expression in SH-SY5Y cells. Treatment of arsenite caused ß-catenin accumulation in the nucleus. Additionally, arsenite treatment decreased the activity of GSK3, an enzyme that phosphorylates and targets ß-catenin for degradation by proteasome, without activation of its upstream kinase, Akt. Inhibition of PI3K/Akt which negatively regulates GSK3 activity by LY294002 resulted in a decrease in arsenite-mediated ß-catenin nuclear accumulation, and VEGF expression. These results suggested that ß-catenin plays a role in arsenite-induced VEGF in SH-SY5Y cells, and the induction of ß-catenin by arsenite is mediated by inhibition of GSK3 without activating its upstream kinase Akt.


Asunto(s)
Arsenitos/toxicidad , Neuroblastoma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cromonas/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/metabolismo
15.
Food Chem Toxicol ; 59: 129-36, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756170

RESUMEN

Glyphosate is an active ingredient of the most widely used herbicide and it is believed to be less toxic than other pesticides. However, several recent studies showed its potential adverse health effects to humans as it may be an endocrine disruptor. This study focuses on the effects of pure glyphosate on estrogen receptors (ERs) mediated transcriptional activity and their expressions. Glyphosate exerted proliferative effects only in human hormone-dependent breast cancer, T47D cells, but not in hormone-independent breast cancer, MDA-MB231 cells, at 10⁻¹² to 10⁻6M in estrogen withdrawal condition. The proliferative concentrations of glyphosate that induced the activation of estrogen response element (ERE) transcription activity were 5-13 fold of control in T47D-KBluc cells and this activation was inhibited by an estrogen antagonist, ICI 182780, indicating that the estrogenic activity of glyphosate was mediated via ERs. Furthermore, glyphosate also altered both ERα and ß expression. These results indicated that low and environmentally relevant concentrations of glyphosate possessed estrogenic activity. Glyphosate-based herbicides are widely used for soybean cultivation, and our results also found that there was an additive estrogenic effect between glyphosate and genistein, a phytoestrogen in soybeans. However, these additive effects of glyphosate contamination in soybeans need further animal study.


Asunto(s)
Neoplasias de la Mama/inducido químicamente , Disruptores Endocrinos/toxicidad , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Glicina/análogos & derivados , Herbicidas/toxicidad , Proteínas de Neoplasias/agonistas , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Disruptores Endocrinos/química , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Estrógenos/química , Estrógenos/toxicidad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Genisteína/farmacología , Glicina/antagonistas & inhibidores , Glicina/toxicidad , Herbicidas/antagonistas & inhibidores , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Hormono-Dependientes/inducido químicamente , Neoplasias Hormono-Dependientes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Glifosato
16.
Cell Mol Biol Lett ; 18(1): 58-74, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23161404

RESUMEN

Glycogen synthase kinase-3 (GSK3) and p53 play crucial roles in the mitochondrial apoptotic pathway and are known to interact in the nucleus. However, it is not known if GSK3 has a regulatory role in the mitochondrial translocation of p53 that participates in apoptotic signaling following DNA damage. In this study, we demonstrated that lithium and SB216763, which are pharmacological inhibitors of GSK3, attenuated p53 accumulation and caspase-3 activation, as shown by PARP cleavage induced by the DNA-damaging agents doxorubicin, etoposide and camptothecin. Furthermore, each of these agents induced translocation of p53 to the mitochondria and activated the mitochondrial pathway of apoptosis, as evidenced by the release of cytochrome C from the mitochondria. Both mitochondrial translocation of p53 and mitochondrial release of cytochrome C were attenuated by inhibition of GSK3, indicating that GSK3 promotes the DNA damage-induced mitochondrial translocation of p53 and the mitochondrial apoptosis pathway. Interestingly, the regulation of p53 mitochondrial translocation by GSK3 was only evident with wild-type p53, not with mutated p53. GSK3 inhibition also reduced the phosphorylation of wild-type p53 at serine 33, which is induced by doxorubicin, etoposide and camptothecin in the mitochondria. Moreover, inhibition of GSK3 reduced etoposide-induced association of p53 with Bcl2 and Bax oligomerization. These findings show that GSK3 promotes the mitochondrial translocation of p53, enabling its interaction with Bcl2 to allow Bax oligomerization and the subsequent release of cytochrome C. This leads to caspase activation in the mitochondrial pathway of intrinsic apoptotic signaling.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Neuroblastoma/genética , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/genética , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Daño del ADN , Doxorrubicina/farmacología , Etopósido/farmacología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo
17.
Chem Res Toxicol ; 25(10): 2194-202, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22971010

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is a critical regulator of cellular responses to hypoxia. Under normoxic conditions, the cellular HIF-1α level is regulated by hydroxylation by prolyl hydroxylases (PHDs), ubiquitylation, and proteasomal degradation. During hypoxia, degradation decreases, and its intracellular level is increased. Exogenously administered nitric oxide (NO)-donor drugs stabilize HIF-1α; thus, NO is suggested to mimic hypoxia. However, the role of low levels of endogenously produced NO generated during hypoxia in HIF-1α stabilization has not been defined. Here, we demonstrate that NO and reactive oxygen species (ROS) produced endogenously by human colon carcinoma HCT116 cells are responsible for HIF-1α accumulation in hypoxia. The antioxidant N-acetyl-L-cysteine (NAC) and NO synthase inhibitor N(G)-monomethyl L-arginine (L-NMMA) effectively reduced HIF-1α stabilization and decreased HIF-1α hydroxylation. These effects suggested that endogenous NO and ROS impaired PHD activity, which was confirmed by reversal of L-NMMA- and NAC-mediated effects in the presence of dimethyloxaloylglycine, a PHD inhibitor. Thiol reduction with dithiothreitol decreased HIF-1α stabilization in hypoxic cells, while dinitrochlorobenzene, which stabilizes S-nitrosothiols, favored its accumulation. This suggested that ROS- and NO-mediated HIF-1α stabilization involved S-nitrosation, which was confirmed by demonstrating increased S-nitrosation of PHD2 during hypoxia. Our results support a regulatory mechanism of HIF-1α during hypoxia in which endogenously generated NO and ROS promote inhibition of PHD2 activity, probably by its S-nitrosation.


Asunto(s)
Neoplasias del Colon/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hipoxia de la Célula , Colon/citología , Colon/metabolismo , Colon/patología , Neoplasias del Colon/patología , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Nitrosación , Procolágeno-Prolina Dioxigenasa/metabolismo
18.
Toxicol In Vitro ; 26(3): 386-95, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22244921

RESUMEN

Vascular endothelium has been considered as a target for arsenic-induced cardiovascular toxicity. The present study demonstrated that arsenite caused slow and sustained elevation of intracellular free calcium levels ([Ca2+]i) in HMEC-1, a human microvessel-derived endothelial cell line, in a concentration-dependent manner. Pretreatment with U-73122 (a specific PLC inhibitor) or 2-APB (a specific IP3 receptor antagonist) attenuated this effect, suggesting that PLC/IP3 signaling cascade is involved in arsenite-induced elevation of [Ca2+]i. Cytotoxic concentrations of arsenite (5 and 10 µM) significantly enhanced endothelial nitric oxide synthase (eNOS) phosphorylation, nitric oxide (NO) production and apoptosis after 24-h exposure. Additionally, 2-APB attenuated eNOS phosphorylation and apoptosis induced by arsenite, indicating that Ca2+ -mediated eNOS activation participates in arsenite-induced endothelial cell apoptosis. Moreover, we also found that non-apoptotic concentrations of arsenite (0.5 and 1 µM) dramatically mitigated thrombin-induced rapid transient rise of [Ca2+]i, eNOS phosphorylation and NO production, suggesting functional disruption of endothelial by arsenite, and these effects occurred without an alteration of PLC-ß1 and thrombin receptor levels. Altogether, the results reveal that arsenite induces apoptotic cell death and endothelial dysfunction as indicated by the reduction of thrombin responses, particularly related to an alteration of intracellular Ca2+ homeostasis.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenitos/toxicidad , Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Compuestos de Sodio/toxicidad , Arsenitos/administración & dosificación , Compuestos de Boro/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/patología , Estrenos/farmacología , Humanos , Microvasos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Pirrolidinonas/farmacología , Transducción de Señal/efectos de los fármacos , Compuestos de Sodio/administración & dosificación , Trombina/metabolismo
19.
Environ Toxicol Pharmacol ; 33(1): 53-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22120617

RESUMEN

Arsenic widely contaminates the environment, especially in drinking water. Although it is a known carcinogen in humans, its carcinogenic mechanism has not yet been clarified. Here, we demonstrated that a low concentration of arsenite treatment induced proliferation of human neuroblastoma SH-SY5Y cells as indicated by increases in cell viability and BrdU incorporation. Additionally, arsenite increased VEGF expression and secretion. Inhibition of VEGF-induced signaling by SU4312, the inhibitor of VEGF receptor 2 kinase, and by treatment with anti-VEGF antibody blocked arsenite-induced increases in cell proliferation. Moreover, arsenite caused activation of ERK, a key signaling molecule involved in cell proliferation, and this activation was attenuated by SU4312, suggesting that ERK activation contributes to VEGF-mediated cell proliferation induced by arsenite. Collectively, the present study reveals that a mechanism underlying arsenic-induced cell proliferation may be through induction and activation of VEGF signaling, and this may subsequently contribute to tumor formation.


Asunto(s)
Arsenitos/toxicidad , Proliferación Celular/efectos de los fármacos , Neuroblastoma/patología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fase S/efectos de los fármacos
20.
Environ Toxicol Pharmacol ; 31(1): 119-28, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21787676

RESUMEN

The cholinergic system in lymphocytes is hypothesized to be a key target for neurotoxic organophosphates (OPs). The present study determined the comparative effects of paraoxon, the active metabolite of OP-parathion, which is detected in the human neuroblastoma line, SH-SY5Y, and leukemic T-lymphocytes, MOLT-3, in vitro. Paraoxon induced cytotoxic effects in a dose- and time-dependent manner in both cells. Further, the paraoxon-induced modulatory effects were comparable despite different cell types, including over-expression of N-terminus acetylcholinesterase (N-AChE) protein, a marker of apoptosis, down-regulations of mRNA encoding M1, M2, and M3 muscarinic acetylcholine receptors (mAChRs), and induction in expression of c-Fos gene, an indication of certain mAChR subtype(s) activation. Furthermore, the non-selective cholinergic antagonist atropine partially attenuated the paraoxon-induced N-AChE and c-Fos activations in both types of cells. These results provide initial and additional information that OPs may similarly induce neuro- and immuno-toxic effects through mAChRs activation, and they underline the potential of using lymphocytes for assessing OPs-induced neurotoxicity.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Linfocitos/efectos de los fármacos , Neuronas/efectos de los fármacos , Paraoxon/toxicidad , Sistema Nervioso Parasimpático/citología , Sistema Nervioso Parasimpático/efectos de los fármacos , Acetilcolinesterasa/metabolismo , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colorantes , Genes fos/efectos de los fármacos , Humanos , Indicadores y Reactivos , Agonistas Muscarínicos/toxicidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Sales de Tetrazolio , Tiazoles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...