Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Arch Toxicol ; 80(9): 580-604, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16598496

RESUMEN

To develop and validate a practical, in vitro, cell-based model to assess human hepatotoxicity potential of drugs, we used the new technology of high content screening (HCS) and a novel combination of critical model features, including (1) use of live, human hepatocytes with drug metabolism capability, (2) preincubation of cells for 3 days with drugs at a range of concentrations up to at least 30 times the efficacious concentration or 100 microM, (3) measurement of multiple parameters that were (4) morphological and biochemical, (5) indicative of prelethal cytotoxic effects, (6) representative of different mechanisms of toxicity, (7) at the single cell level and (8) amenable to rapid throughput. HCS is based on automated epifluorescence microscopy and image analysis of cells in a microtiter plate format. The assay was applied to HepG2 human hepatocytes cultured in 96-well plates and loaded with four fluorescent dyes for: calcium (Fluo-4 AM), mitochondrial membrane potential (TMRM), DNA content (Hoechst 33,342) to determine nuclear area and cell number and plasma membrane permeability (TOTO-3). Assay results were compared with those from 7 conventional, in vitro cytotoxicity assays that were applied to 611 compounds and shown to have low sensitivity (<25%), although high specificity ( approximately 90%) for detection of toxic drugs. For 243 drugs with varying degrees of toxicity, the HCS, sublethal, cytotoxicity assay had a sensitivity of 93% and specificity of 98%. Drugs testing positive that did not cause hepatotoxicity produced other serious, human organ toxicities. For 201 positive assay results, 86% drugs affected cell number, 70% affected nuclear area and mitochondrial membrane potential and 45% affected membrane permeability and 41% intracellular calcium concentration. Cell number was the first parameter affected for 56% of these drugs, nuclear area for 34% and mitochondrial membrane potential for 29% and membrane permeability for 7% and intracellular calcium for 10%. Hormesis occurred for 48% of all drugs with positive response, for 26% of mitochondrial and 34% nuclear area changes and 12% of cell number changes. Pattern of change was dependent on the class of drug and mechanism of toxicity. The ratio of concentrations for in vitro cytotoxicity to maximal efficaciousness in humans was not different across groups (12+/-22). Human toxicity potential was detected with 80% sensitivity and 90% specificity at a concentration of 30x the maximal efficacious concentration or 100 microM when efficaciousness was not considered. We conclude that human hepatotoxicity is highly concordant with in vitro cytotoxicity in this novel model and as detected by HCS.


Asunto(s)
Alternativas a las Pruebas en Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Hepatocitos/efectos de los fármacos , Pruebas de Toxicidad/métodos , Xenobióticos/toxicidad , Calcio/metabolismo , Carcinoma Hepatocelular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Concentración 50 Inhibidora , Neoplasias Hepáticas , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Xenobióticos/clasificación
2.
Toxicol Lett ; 123(1): 33-41, 2001 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-11514103

RESUMEN

Methylmercury (MeHg) is a toxicant that targets the kidney among other tissues. MeHg accumulates in the kidney, where it indirectly produces oxidative stress due to glutathione depletion and leakage of reactive oxygen species from the mitochondria. Glutathione is believed to have an important role in protecting the kidney against MeHg toxicity, and MeHg exposure is known to result in the induction of GSH synthesis through the upregulation of the enzyme glutamate-cysteine ligase (GLCL). GLCL, the rate-limiting enzyme in GSH synthesis, is composed of two subunits, a large catalytic (GLCLc) and a smaller regulatory (GLCLr) subunit. In this study we show that GLCLc and GLCLr mRNAs and GLCLc protein are localized in the paracortical region of the mouse kidney, the area of the kidney with the highest MeHg concentration, and that the upregulation of these mRNAs induced by MeHg is also located to the same region. This supports the role of GLCL in protection against MeHg toxicity in the kidney.


Asunto(s)
Glutamato-Cisteína Ligasa/biosíntesis , Riñón/efectos de los fármacos , Compuestos de Metilmercurio/toxicidad , ARN Mensajero/biosíntesis , Animales , Femenino , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Riñón/enzimología , Corteza Renal/efectos de los fármacos , Corteza Renal/enzimología , Médula Renal/efectos de los fármacos , Médula Renal/enzimología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos
3.
Toxicol Lett ; 122(2): 119-29, 2001 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-11439218

RESUMEN

Glutamate-cysteine ligase (GLCL), the rate-limiting enzyme in glutathione (GSH) synthesis is composed of two subunits, a catalytic (GLCLc) and a regulatory subunit (GLCLr). These two subunits are known to be differentially regulated in vitro, in different cell types and in response to various xenobiotic exposures. In this study, we examined whether these two subunits can also be differentially regulated in vivo. We found that GLCLc and GLCLr are differentially regulated at the transcriptional level in a tissue-dependent manner in female mice treated with methylmercury (MeHg). MeHg caused a downregulation of both subunit mRNAs in the liver, upregulation of both subunit mRNAs in the kidney and upregulation of only the catalytic subunit mRNA in the small intestine of female mice treated with a single dose of MeHg (6 mg/kg) by intraperitoneal injection. These results suggest that GLCLc and GLCLr can be differentially regulated in vivo, and that this regulation is tissue dependent in the mouse.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glutamato-Cisteína Ligasa/genética , Compuestos de Metilmercurio/toxicidad , ARN Mensajero/análisis , Animales , Femenino , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/análisis , Glutatión/metabolismo , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos
4.
Toxicol Sci ; 57(1): 141-6, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10966520

RESUMEN

The antioxidant tripeptide glutathione has been proposed to be important in defense against oxidative stress and heavy metal toxicity. We evaluated alterations in glutathione regulation and synthesis associated with low-level chronic methylmercury (MeHg) exposure in the developing mouse fetus. Female C57Bl/6 mice were given 0, 3, or 10 ppm MeHg in the drinking water for 2 weeks prior to breeding and throughout pregnancy. Fetuses were collected on gestational days (gd) 12 and 16. Total glutathione, reduced glutathione (GSH), oxidized glutathione (GSSR), and glutamate-L-cysteine ligase (Glcl) activity were assessed in yolk sacs and fetuses at gd 16. Western and Northern blots for Glcl-catalytic (Glclc) and Glcl-regulatory (Glclr) subunits were performed on gd 12 and gd 16 fetuses. There were no changes in total glutathione in gd 16 mouse fetuses with exposure, but there were dose-related decreases in GSH and increases in GSSR. In contrast, visceral yolk sacs exhibited an increase in total glutathione in the low-dose groups, but no changes in the high-dose group. There were no changes in Glcl activity in fetuses, but there was a 2-fold increase in Glcl activity in yolk sacs from both low-dose and high-dose groups. There was a 2-fold induction in GLCLC: mRNA and protein in the gd 16 yolk sacs at both 3 and 10 ppm MeHg. No treatment-related changes in Glclr protein in either gd 12 or gd 16 yolk sacs or fetuses were found. Thus, the yolk sac is capable of up-regulating Glclc and GSH synthetic capacity in response to MeHg exposure. This increase appears to be sufficient to resist MeHg-induced GSH depletion in the yolk sac; however fetal glutathione redox status is compromised with exposure to 10 ppm MeHg.


Asunto(s)
Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Compuestos de Metilmercurio/toxicidad , Animales , Northern Blotting , Western Blotting , Cromatografía Líquida de Alta Presión , Femenino , Feto/metabolismo , Mercurio/metabolismo , Compuestos de Metilmercurio/farmacocinética , Ratones , Ratones Endogámicos C57BL , Embarazo , Saco Vitelino/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 278(4): L696-702, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10749746

RESUMEN

The development of drug resistance of tumors is multifactorial and still poorly understood. Some cytotoxic drugs generate free radicals, and, therefore, antioxidant enzymes may contribute to drug resistance. We investigated the levels of manganese superoxide dismutase (Mn SOD), its inducibility, and its protective role against tumor necrosis factor-alpha and cytotoxic drugs (cisplatin, epirubicin, methotrexate, and vindesin) in human pleural mesothelioma (M14K) and pulmonary adenocarcinoma (A549) cells. We also studied other major antioxidant mechanisms in relation to oxidant and drug resistance of these cells. A549 cells were more resistant than M14K cells toward both oxidants (hydrogen peroxide and menadione) and all the cytotoxic drugs tested. M14K cells contained higher basal Mn SOD activity than A549 cells (28.3 +/- 3.4 vs. 1.8 +/- 0.3 U/mg protein), and Mn SOD activity was significantly induced by tumor necrosis factor-alpha only in A549 cells (+524%), but the induction did not offer any protection during subsequent oxidant or drug exposure. Mn SOD was not induced significantly in either of these cell lines by any of the cytotoxic drugs (0.007-2 microM, 48 h) tested when assessed by Northern blotting, Western blotting, or specific activity. A549 cells contained higher catalase activity than M14K cells (7.6 +/- 1.3 vs. 3.6 +/- 0.5 nmol O(2). min(-1). mg protein(-1)). They also contained twofold higher levels of glutathione and higher immunoreactivity of the heavy subunit of gamma-glutamylcysteine synthetase than M14K cells. Experiments with inhibitors of gamma-glutamylcysteine synthetase and catalase supported our conclusion that mechanisms associated with glutathione contribute to the drug resistance of these cells.


Asunto(s)
Adenocarcinoma/metabolismo , Antioxidantes/metabolismo , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Adenocarcinoma/patología , Catalasa/metabolismo , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares/patología , Mesotelioma/patología , Oxidantes/farmacología , ARN Mensajero/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Células Tumorales Cultivadas , Vitamina K/farmacología
7.
Neurotoxicology ; 20(5): 793-804, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10591515

RESUMEN

Previous studies indicate that the ability of cells to up-regulate levels of intracellular glutathione (GSH) synthesis may determine their sensitivity to MeHg exposure. The purpose of the current study is two-fold. First, we determined whether the vulnerability of the developing central nervous system (CNS) to MeHg lies in its intracellular GSH content. The intracellular GSH content and the activity of gamma-glutamyl cysteine synthetase (GCS) were determined with and without MeHg exposure in primary cultures of rat embryonic CNS cells. In addition, the effect of GSH modulation on MeHg-induced cytotoxicity was determined. Second, we characterized the mechanism of GCS regulation, initially by studying the GCS heavy chain subunit (GCS-HC). Primary embryonic limb bud cells were used as a reference cell type for comparing the response of CNS cells. The results indicate that constitutive intracellular GSH content, GCS activity, and GCS-HC mRNA and protein levels of CNS cells were approximately ten-, two-, five-, and ten-fold higher, respectively, than those in limb bud cells. A dose-dependent increase in GSH levels and GCS activity was observed in CNS and limb bud cells following 1 and 2 microM MeHg exposure for 20 hr. Further characterization of GCS up-regulation in CNS cells showed that the increase in GCS activity following MeHg exposure, unlike limb bud cells, was not accompanied by an elevation of GCS-HC mRNA and protein levels. Pretreatment with N-acetylcysteine led to a significant increase in intracellular GSH, while L-buthionine-(S,R)-sulfoximine (BSO) resulted in decreased GSH levels, however neither pretreatment had a significant impact on MeHg-induced cytotoxicity in either cell type. Our results suggest that although oxidative stress may mediate aspects of MeHg toxicity, disruption of GSH homeostasis alone is not responsible for the sensitivity of embryonic CNS cells to MeHg.


Asunto(s)
Sistema Nervioso Central/patología , Embrión de Mamíferos/citología , Glutatión/fisiología , Compuestos de Metilmercurio/toxicidad , Neuronas/efectos de los fármacos , Acetilcisteína/farmacología , Aminoaciltransferasas/biosíntesis , Animales , Butionina Sulfoximina/farmacología , Diferenciación Celular/efectos de los fármacos , Sistema Nervioso Central/embriología , Embrión de Mamíferos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Extremidades/embriología , Extremidades/inervación , Citometría de Flujo , Depuradores de Radicales Libres/farmacología , Homeostasis/fisiología , Neuronas/patología , Estrés Oxidativo/fisiología , Pirazoles , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
8.
Toxicol Lett ; 110(1-2): 1-9, 1999 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-10593589

RESUMEN

Methylmercury (MeHg) is widely known for its potent neurotoxic properties. One proposed mechanism of action of MeHg relates to its high affinity for sulfhydryl groups, especially those found on glutathione (GSH) and proteins. Previous studies have shown that acute MeHg exposure results in an increase in the mRNA for the rate-limiting enzyme in GSH synthesis, glutamate-cysteine ligase (GLCL) (also known as gamma-glutamylcysteine synthetase). In this study, we evaluated the effects of subchronic (12-week) MeHg exposure at 0, 3 or 10 ppm in the drinking water on GSH levels, GLCL catalytic (GLCLC) and regulatory subunit mRNA and protein levels, and GLCL activity in brain, liver and kidney tissue of C57B1/6 female mice. Contrary to previous findings in rats, there were no changes in GSH concentration in any of the tissues examined. However, there was an increase in GLCLC protein in the brain, which was accompanied by a 30% increase in GLCL activity. We conclude that up-regulation of GSH synthetic capacity in the brains of mice is a sensitive biomarker of subchronic MeHg exposure.


Asunto(s)
Encéfalo/efectos de los fármacos , Glutamato-Cisteína Ligasa/biosíntesis , Compuestos de Metilmercurio/toxicidad , Animales , Northern Blotting , Western Blotting , Encéfalo/enzimología , Cromatografía Líquida de Alta Presión , Inducción Enzimática , Femenino , Glutatión/metabolismo , Riñón/efectos de los fármacos , Riñón/enzimología , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
9.
Ann Oncol ; 10(11): 1361-7, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10631466

RESUMEN

BACKGROUND: Etoposide mediates its cytotoxicity by inducing apoptosis. Thus, mechanisms which regulate apoptosis should also affect drug resistance. Oxidants and antioxidants have been shown to participate in the regulation of apoptosis. We were interested in studying whether responsiveness of acute myeloblastic leukemia (AML) cells to etoposide is mediated by oxidative stress and glutathione levels. PATIENTS AND METHODS: Two subclones of the OCI/AML-2 cell line which are etoposide-sensitive (ES), and etoposide-resistant (ER), were established by the authors at the University of Oulu, and used as models. Assays for apoptosis included externalization of phosphatidylserine (as evidenced by annexin V binding), and caspase activation as indicated by cleavage of poly(ADP-ribose)polymerase (Western blotting). Peroxide formation was analyzed by flow cytometry. Glutathione and gamma-glutamylcysteine synthetase (gamma-GCS) levels were determined spectrophotometrically and by Western blotting, respectively. RESULTS: Etoposide-induced apoptosis was evident 12 hours after treatment in the ES subclone, but was apparent in the ER subclone only after 24 hours. The basal glutathione and gamma-GCS levels were higher in the ER than the ES subclone. Etoposide increased peroxide formation in both subclones after 12-hour exposure. Significant depletion of glutathione was observed in the ES subclone during etoposide exposure, while glutathione levels were maintained in the ER subclone. In neither of the subclones was induction of gamma-GCS observed during 24-hour exposure to etoposide. Furthermore, the catalytic subunit of gamma-GCS was cleaved during apoptosis, concurrent with depletion of intracellular glutathione. When glutathione was depleted by treatment with buthionine sulfoximine, a direct inhibitor of gamma-GCS, the sensitivity to etoposide was increased, particularly in the ER subclone. CONCLUSIONS: The results underline the significance of glutathione biosynthesis in the responsiveness of AML cells to etoposide. The molecular mechanisms mediating glutathione depletion during etoposide exposure might include the cleavage of the catalytic subunit of gamma-GCS.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/fisiología , Etopósido/farmacología , Glutamato-Cisteína Ligasa/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Western Blotting , Resistencia a Antineoplásicos , Citometría de Flujo , Glutamato-Cisteína Ligasa/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Sensibilidad y Especificidad , Células Tumorales Cultivadas
10.
Environ Health Perspect ; 106 Suppl 5: 1179-84, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9788895

RESUMEN

Phosphotyrosine phosphatases (PTPs) serve as important regulators of cellular signal transduction pathways. PTPs are sensitive targets of oxidative stress and may be inhibited by treatments that induce intracellular oxidation. The effects of PTP inactivation under oxidizing conditions are amplified by the redox-linked activation of key protein tyrosine kinases (PTKs), thus leading to the initiation of phosphotyrosine-signaling cascades that are no longer under normal receptor control. These ligand-independent signals result in the accumulation of protein phosphotyrosine, the generation of second messengers, the activation of downstream kinases, and the nuclear translocation of nuclear factor kappa B (NF-kappa B). In this review we consider the relative contribution of oxidative stress to the effects of PTP inhibition by vanadium-based compounds in lymphocytes. Although the inactivation of PTPs can lead to NF-kappa B mobilization in the presence of antioxidants, the other effects noted appear to require a threshold of intracellular oxidation. The combined effects of oxidative stress on signal transduction cascades reflect a synergy between the initiation of signals by PTKs and the loss of control by PTPs. This suggests a mechanism by which environmental agents that cause oxidative stress may alter the course of cellular responses through induction or enhancement of signaling cascades leading to functional changes or cell death.


Asunto(s)
Proteínas Tirosina Fosfatasas/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/enzimología , Linfocitos/inmunología , Estrés Oxidativo , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Vanadatos/farmacología
11.
J Biol Chem ; 272(17): 11541-9, 1997 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-9111069

RESUMEN

The role of intracellular oxidative stress in the mechanism of action of phosphotyrosine phosphatase (PTP) inhibitors was studied using three vanadium-based compounds. Sodium orthovanadate (Na3VO4), sodium oxodiperoxo(1,10-phenanthroline)vanadate(V) (pV(phen), and bis(maltolato)-oxovanadium(IV) (BMOV) differentially induced oxidative stress in lymphocytes. Treatment with pV(phen), which caused intracellular oxidation, induced strong protein tyrosine phosphorylation compared with Na3VO4 and BMOV. Syk family kinases and the mitogen-activated protein kinase erk2 were rapidly activated by pV(phen) but not by BMOV or Na3VO4. In contrast, both BMOV and pV(phen) strongly activated NF-kappaB. The antioxidant pyrrolidine dithiocarbamate (PDTC) greatly diminished the intracellular oxidation and protein phosphotyrosine accumulation induced by pV(phen). Pretreatment of cells with PDTC reduced and delayed the activation of Syk kinases and erk2. However, NF-kappaB activation by pV(phen) was markedly enhanced in lymphocytes pretreated with PDTC, and another antioxidant, N-acetylcysteine, did not prevent the activation of NF-kappaB by BMOV. These results indicate a role for oxidative stress in the biological effects of some PTP inhibitors, whereas NF-kappaB activation by PTP inhibitors is mediated by mechanisms independent of intracellular redox status.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas I-kappa B , Linfocitos/fisiología , Compuestos Organometálicos/farmacología , Estrés Oxidativo/fisiología , Fenantrolinas/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Antioxidantes/farmacología , Linfocitos B/fisiología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Humanos , Antígenos Comunes de Leucocito/efectos de los fármacos , Inhibidor NF-kappaB alfa , Oxidación-Reducción , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Pironas/farmacología , Transducción de Señal , Linfocitos T/fisiología , Vanadatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA