Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Nat Med ; 5(5): 582-5, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10229238

RESUMEN

The sedative drug thalidomide ([+]-alpha-phthalimidoglutarimide), once abandoned for causing birth defects in humans, has found new therapeutic license in leprosy and other diseases, with renewed teratological consequences. Although the mechanism of teratogenesis and determinants of risk remain unclear, related teratogenic xenobiotics are bioactivated by embryonic prostaglandin H synthase (PHS) to a free-radical intermediates that produce reactive oxygen species (ROS), which cause oxidative damage to DNA and other cellular macromolecules. Similarly, thalidomide is bioactivated by horseradish peroxidase, and oxidizes DNA and glutathione, indicating free radical-mediated oxidative stress. Furthermore, thalidomide teratogenicity in rabbits is reduced by the PHS inhibitor acetylsalicylic acid, indicating PHS-catalyzed bioactivation. Here, we show in rabbits that thalidomide initiates embryonic DNA oxidation and teratogenicity, both of which are abolished by pre-treatment with the free radical spin trapping agent alpha-phenyl-N-t-butylnitrone (PBN). In contrast, in mice, a species resistant to thalidomide teratogenicity, thalidomide does not enhance DNA oxidation, even at a dose 300% higher than that used in rabbits, providing insight into an embryonic determinant of species-dependent susceptibility. In addition to their therapeutic implications, these results constitute direct evidence that the teratogenicity of thalidomide may involve free radical-mediated oxidative damage to embryonic cellular macromolecules.


Asunto(s)
Daño del ADN , Embrión de Mamíferos/metabolismo , Hipnóticos y Sedantes/metabolismo , Deformidades Congénitas de las Extremidades/etiología , Teratógenos/metabolismo , Talidomida/metabolismo , Animales , Óxidos N-Cíclicos , Resistencia a Medicamentos , Pérdida del Embrión , Embrión de Mamíferos/patología , Femenino , Depuradores de Radicales Libres/farmacología , Radicales Libres , Hernia Umbilical , Hipnóticos y Sedantes/efectos adversos , Ratones , Óxidos de Nitrógeno/farmacología , Oxidación-Reducción , Embarazo , Conejos , Especificidad de la Especie , Talidomida/efectos adversos
2.
Oncogene ; 26(20): 2914-24, 2007 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-17072338

RESUMEN

Previously, we showed that Src tyrosine kinases are activated early in the development of human colon cancer and are suppressed as intestinal cells differentiate. We identified RACK1 as an endogenous substrate, binding partner and inhibitor of Src. Here we show (by overexpressing RACK1, depleting Src or RACK1 and utilizing cell-permeable peptides that perturb RACK1's interaction with Src) that RACK1 regulates growth of colon cells by suppressing Src activity at G(1) and mitotic checkpoints, and consequently delaying cell cycle progression. Activated Src rescues RACK1-inhibited growth of HT-29 cells. Conversely, inhibiting Src abolishes growth promoted by RACK1 depletion in normal cells. Two potential mechanisms whereby RACK1 regulates mitotic exit are identified: suppression of Src-mediated Sam68 phosphorylation and maintenance of the cyclin-dependent kinase (CDK) 1-cyclin B complex in an active state. Our results reveal novel mechanisms of cell cycle control in G(1) and mitosis of colon cells. The significance of this work lies in the discovery of a mechanism by which the growth of colon cancer cells can be slowed, by RACK1 suppression of an oncogenic kinase at critical cell cycle checkpoints. Small molecules that mimic RACK1 function may provide a powerful new approach to the treatment of colon cancer.


Asunto(s)
Carcinoma/patología , Ciclo Celular/genética , Proliferación Celular , Neoplasias del Colon/patología , Proteínas de Unión al GTP/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptores de Superficie Celular/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al GTP/metabolismo , Genes cdc/fisiología , Humanos , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Receptores de Cinasa C Activada , Receptores de Superficie Celular/metabolismo , Células Tumorales Cultivadas
3.
Mutat Res ; 396(1-2): 65-78, 1997 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-9434860

RESUMEN

The teratogenicity of many xenobiotics is thought to depend at least in part upon their bioactivation by embryonic cytochromes P450, prostaglandin H synthase (PHS) and lipoxygenases (LPOs) to electrophilic and/or free radical reactive intermediates that covalently bind to or oxidize cellular macromolecules such as DNA, protein and lipid, resulting in in utero death or teratogenesis. Using as models the tobacco carcinogens benzo[a]pyrene (B[a]P) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), the anticonvulsant drug phenytoin, structurally related anticonvulsants (e.g. mephenytoin, nirvanol, trimethadione, dimethadione) and the sedative drug thalidomide, we have examined the potential teratologic relevance of free radical-initiated, reactive oxygen species (ROS)-mediated oxidative molecular target damage, genotoxicity (micronucleus formation) and DNA repair in mouse and rabbit models in vivo and in embryo culture, and in vitro using purified enzymes or cultured rat skin fibroblasts. These teratogens were bioactivated by PHS and LPOs to free radical reactive intermediary metabolites, characterized by electron spin resonance spectrometry, that initiated ROS formation, including hydroxyl radicals, which were characterized by salicylate hydroxylation. ROS-initiated oxidation of DNA (8-hydroxy-2'-deoxyguanosine formation), protein (carbonyl formation), glutathione (GSH) and lipid (peroxidation), and embryotoxicity were shown for phenytoin, its major hydroxylated metabolite 5-(p-hydroxyphenyl)-5-phenylhydantoin [HPPH], thalidomide, B[a]P and NNK in vivo and/or in embryo culture, the latter indicating a teratologically critical role for embryonic, as distinct from maternal, processes. DNA oxidation and teratogenicity of phenytoin and thalidomide were reduced by PHS inhibitors. Oxidative macromolecular lesions and teratogenicity also were reduced by the free radical trapping agent phenylbutylnitrone (PBN), and the antioxidants caffeic acid and vitamin E. In embryo culture, addition of superoxide dismutase (SOD) to the medium enhanced embryonic SOD activity, and SOD or catalase blocked the oxidative lesions and embryotoxicity initiated by phenytoin and B[a]P, suggesting a major contribution of ROS, as distinct from covalent binding, to the teratologic mechanism. In in vivo studies, other antioxidative enzymes like GSH peroxidase, GSH reductase and glucose-6-phosphate dehydrogenase (G6PD) were similarly protective. Even untreated G6PD-deficient mice had enhanced embryopathies, indicating a teratological role for endogenous oxidative stress. In cultured fibroblasts, B[a]P, NNK, phenytoin and HPPH initiated DNA oxidation and micronucleus formation, which were inhibited by SOD. Oxidation of DNA may be particularly critical, since transgenic mice with +/- or -/- deficiencies in the p53 tumor suppressor gene, which facilitates DNA repair, are more susceptible to phenytoin and B[a]P teratogenicity. Even p53-deficient mice treated only with normal saline showed enhanced embryopathies, suggesting the teratological importance of endogenous oxidative stress, as observed with G6PD deficiency. These results suggest that oxidative macromolecular damage may play a role in the teratologic mechanism of xenobiotics that are bioactivated to a reactive intermediate, as well in the mechanism of embryopathies occurring in the absence of xenobiotic exposure.


Asunto(s)
Teratógenos/farmacología , Xenobióticos/toxicidad , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/prevención & control , Animales , Anticonvulsivantes/toxicidad , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Benzo(a)pireno/toxicidad , Biotransformación , Daño del ADN , Desarrollo Embrionario y Fetal/efectos de los fármacos , Radicales Libres , Ratones , Nitrosaminas/toxicidad , Estrés Oxidativo , Fenitoína/toxicidad , Proteínas/efectos de los fármacos , Conejos , Ratas , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos , Teratógenos/farmacocinética , Talidomida/toxicidad , Xenobióticos/farmacocinética
4.
J Biol Chem ; 273(39): 25079-88, 1998 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-9737965

RESUMEN

Phenytoin and related xenobiotics can be bioactivated by embryonic prostaglandin H synthase (PHS) to a teratogenic free radical intermediate. The mechanism of free radical formation was evaluated using photolytic oxidation with sodium persulfate and by EPR spectrometry. Characterization of the products by mass spectrometry suggested that phenytoin photolyzes to a nitrogen-centered radical that rapidly undergoes ring opening to form a carbon-centered radical. PHS-1 was incubated with teratogen (phenytoin, mephenytoin, trimethadione, phenobarbital, and major metabolites) or its vehicle and the free radical spin trap alpha-phenyl-N-t-butylnitrone, and incubations were analyzed by EPR spectrometry. There was no alpha-phenyl-N-t-butylnitrone radical adduct in control incubations. For phenytoin, a putative unstable nitrogen-centered radical adduct and a stable carbon-centered radical adduct were detected. Free radical spin adducts also were detected for all other teratogens and metabolites except carbamazepine. The PHS inhibitor eicosatetraynoic acid abolished the free radical EPR signal. Incubation of 2'-deoxyguanosine with phenytoin and PHS-1 resulted in a 5-fold increase in its oxidation to 8-hydroxy-2'-deoxyguanosine. This is the first direct chemical evidence for PHS-catalyzed bioactivation of phenytoin and related teratogens to a free radical intermediate that initiates DNA oxidation, which may constitute a common molecular mechanism of teratologic initiation.


Asunto(s)
Fenitoína/farmacocinética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Teratógenos/farmacocinética , Biotransformación , Catálisis , Desoxiguanosina/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres , Oxidación-Reducción , Fenitoína/química , Fotoquímica , Teratógenos/química
5.
J Pharmacol Exp Ther ; 280(1): 200-9, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8996197

RESUMEN

UDP-Glucuronosyltransferases (UGTs) are important in the elimination of most xenobiotics, including 5-(p-hydroxyphenyl)-5-phenylhydantoin (HPPH), the major, reputedly nontoxic, metabolite of the anticonvulsant drug phenytoin. However, HPPH alternatively may be bioactivated by peroxidases, such as prostaglandin H synthase, to a reactive intermediate that initiates DNA oxidation (reflected by 8-hydroxy-2'-deoxyguanosine), genotoxicity (reflected by micronuclei) and embryopathy. This hypothesis was evaluated in skin fibroblasts cultured from heterozygous (+/j) and homozygous (j/j) UGT-deficient Gunn rats and in mouse embryo culture, with confirmation of direct NG-glucuronidation of phenytoin in Gunn rats in vivo. HPPH (80 microM) increased micronuclei by 2.0-, 4.8- and 4.6-fold in +/+ UGT-normal cells (P = .03) and +/j and j/j UGT-deficient cells (P = .0001), respectively. HPPH-initiated micronucleus formation was increased 3.0- and 3.4-fold in +/j (P = .02) and j/j (P = .04) UGT-deficient cells, respectively, vs. +/+ UGT-normal cells. Micronuclei were not initiated by 10 microM HPPH in +/+ UGT-normal cells but were increased by 4- and 3.8-fold in +/j and j/j UGT-deficient cells (P = .0001), respectively, and were increased 2.7- and 3.0-fold in +/j (P = .007) and j/j (P = .0002) UGT-deficient cells, respectively, vs. +/+ UGT-normal cells. 8-Hydroxy-2'-deoxyguanosine was increased in j/j UGT-deficient but not +/+ UGT-normal cells treated with 80 microM HPPH (P < .05). The embryopathic potency of 80 microM HPPH in embryo culture, reflected by decreases in anterior neuropore closure, turning, yolk sac diameter and crown-rump length (P < .05), was equivalent to that reported for phenytoin. Phenytoin (80 microM) enhanced micronucleus formation 1.7-, 4.4- and 3.8-fold in +/+ cells (P = .03) and +/j and j/j UGT-deficient cells (P = .0001), respectively. Phenytoin-initiated micronucleus formation was increased about 4-fold in both +/j (P = .006) and j/j (P = .009) UGT-deficient cells vs. +/+ UGT-normal cells, providing the first evidence that the bioactivation and oxidative toxicity of phenytoin itself may be avoided by direct N-glucuronidation, which was confirmed by tandem mass spectrometry. These results further indicate that, with UGT deficiencies, HPPH potentially is a potent mediator of phenytoin-initiated genotoxicity and embryopathy, which may be relevant to teratogenesis and other adverse effects of phenytoin.


Asunto(s)
Anticonvulsivantes/toxicidad , ADN/metabolismo , Embrión de Mamíferos/efectos de los fármacos , Glucuronosiltransferasa/fisiología , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Fenitoína/análogos & derivados , Fenitoína/toxicidad , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Femenino , Masculino , Ratones , Técnicas de Cultivo de Órganos , Oxidación-Reducción , Ratas , Ratas Gunn , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA