Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cardiovasc Res ; 97(1): 23-32, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22997158

RESUMEN

AIMS: Several drugs blocking the rapidly activating potassium (K(r)) channel cause malformations (including cardiac defects) and embryonic death in animal teratology studies. In humans, these drugs have an established risk for acquired long-QT syndrome and arrhythmia. Recently, associations between cardiac defects and spontaneous abortions have been reported for drugs widely used in pregnancy (e.g. antidepressants), with long-QT syndrome risk. To investigate whether a common embryonic adverse-effect mechanism exists in the human, rat, and rabbit embryos, we made a comparative study of embryonic cardiomyocytes from all three species. METHODS AND RESULTS: Patch-clamp and quantitative-mRNA measurements of K(r) and slowly activating K (K(s)) channels were performed on human, rat, and rabbit primary cardiomyocytes and cardiac samples from different embryo-foetal stages. The K(r) channel was present when the heart started to beat in all species, but was, in contrast to human and rabbit, lost in rats in late organogenesis. The specific K(r)-channel blocker E-4031 prolonged the action potential in a species- and development-dependent fashion, consistent with the observed K(r)-channel expression pattern and reported sensitive periods of developmental toxicity. E-4031 also increased the QT interval and induced 2:1 atrio-ventricular block in multi-electrode array electrographic recordings of rat embryos. The K(s) channel was expressed in human and rat throughout the embryo-foetal period but not in rabbit. CONCLUSION: This first comparison of mRNA expression, potassium currents, and action-potential characteristics, with and without a specific K(r)-channel blocker in human, rat, and rabbit embryos provides evidence of K(r)-channel inhibition as a common mechanism for embryonic malformations and death.


Asunto(s)
Cardiopatías Congénitas/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/toxicidad , Bloqueadores de los Canales de Potasio/toxicidad , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Piridinas/toxicidad , Teratógenos/toxicidad , Potenciales de Acción , Animales , Bloqueo Atrioventricular/inducido químicamente , Bloqueo Atrioventricular/metabolismo , Células Cultivadas , Canal de Potasio ERG1 , Electrocardiografía , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Humanos , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/metabolismo , Cinética , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/metabolismo , Miocitos Cardíacos/metabolismo , Organogénesis , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , ARN Mensajero/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Especificidad de la Especie
2.
J Neurosci ; 32(48): 17297-305, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197721

RESUMEN

γ-Secretase inhibition represents a major therapeutic strategy for lowering amyloid ß (Aß) peptide production in Alzheimer's disease (AD). Progress toward clinical use of γ-secretase inhibitors has, however, been hampered due to mechanism-based adverse events, primarily related to impairment of Notch signaling. The γ-secretase inhibitor MRK-560 represents an exception as it is largely tolerable in vivo despite displaying only a small selectivity between Aß production and Notch signaling in vitro. In exploring the molecular basis for the observed tolerability, we show that MRK-560 displays a strong preference for the presenilin 1 (PS1) over PS2 subclass of γ-secretases and is tolerable in wild-type mice but causes dose-dependent Notch-related side effect in PS2-deficient mice at drug exposure levels resulting in a substantial decrease in brain Aß levels. This demonstrates that PS2 plays an important role in mediating essential Notch signaling in several peripheral organs during pharmacological inhibition of PS1 and provide preclinical in vivo proof of concept for PS2-sparing inhibition as a novel, tolerable and efficacious γ-secretase targeting strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Encéfalo/efectos de los fármacos , Presenilina-2/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Ratones , Presenilina-2/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
3.
Toxicol Sci ; 121(2): 328-42, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21427059

RESUMEN

Prediction of developmental toxicity in vitro could be based on short-time toxicogenomic endpoints in embryo-derived cell lines. Microarray studies in P19 mouse embryocarcinoma cells and mouse embryos have indicated that valproic acid (VPA), an inducer of neural tube defects, deregulates the expression of many genes, including those critically involved in neural tube development. In this study, we exposed undifferentiated R1 mouse embryonic stem cells to VPA and VPA analogs for 6 h and used CodeLink whole-genome expression microarrays to define VPA-responsive genes correlating with teratogenicity. Compared with the nonteratogenic analog 2-ethyl-4-methylpentanoic acid, VPA and the teratogenic VPA analog (S)-2-pentyl-4-pentynoic acid deregulated a much larger number of genes. Five genes (of ∼2500 array probes correlating with the separation) were sufficient to effectively separate teratogens from nonteratogens. A large fraction of the target genes correlating with teratogenicity are functionally related to embryonic development and morphogenesis, including neural tube formation and closure. Similar responses in R1 were found for most genes previously identified as VPA responsive in P19 and embryos. A subset of target genes was evaluated as candidate markers predictive of potential teratogenicity against a range of known teratogens using TaqMan expression arrays. These marker genes showed a positive predictive value for the teratogens butyrate and trichostatin A, which like VPA and (S)-2-pentyl-4-pentynoic acid are known histone deacetylase (HDAC) inhibitors but not for compounds that are likely to act by other mechanisms. This indicates that HDAC inhibition may be a major mechanism by which VPA induces gene deregulation and possibly teratogenicity.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Ácidos Grasos Insaturados/toxicidad , Regulación del Desarrollo de la Expresión Génica , Ácido Valproico/análogos & derivados , Anomalías Inducidas por Medicamentos/genética , Animales , Butiratos/toxicidad , Línea Celular , Desarrollo Embrionario/genética , Células Madre Embrionarias/metabolismo , Estudios de Asociación Genética , Marcadores Genéticos , Inhibidores de Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/toxicidad , Ratones , Análisis por Micromatrices , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Teratógenos/toxicidad , Toxicogenética , Ácido Valproico/toxicidad
4.
Toxicol Pathol ; 39(2): 325-36, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21270424

RESUMEN

The dual peroxisome-proliferator-activated receptor (PPAR) α/γ agonist tesaglitazar has been shown to produce fibrosarcomas in rats. Here, the authors studied morphology, proliferation, differentiation, and inflammation markers in adipose tissue from rats exposed to 1, 3, or 10 µmol/kg tesaglitazar for 2 or 12 weeks, including recovery groups (12 weeks treatment followed by 12 weeks recovery), and 3 or 10 µmol/kg tesaglitazar for 24 weeks. Subcutaneous white and brown fat revealed reversible dose-related histopathological alterations and after 12 and 24 weeks developed areas of thickened skin (fatty lumps). There was a dose-dependent increase in proliferation of interstitial cells in white and brown fat as shown by increased mitotic index in all dose groups after 2 weeks. This was limited to the high dose after 12 and 24 weeks in white fat. Gene expression analyses showed that while tesaglitazar induced differentiation of adipose tissue characterized with a switch in cyclin D1 and D3 mRNA by 12 weeks, longer exposure at high doses reversed this differentiation concurrent with a reappearance of early adipocyte and inflammatory markers. These data suggest that sustained increased turnover of mesenchymal cells in adipose tissues, concomitant with onset of inflammation and fibrosis, drives development of fibrosarcomas in rats treated with tesaglitazar.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fibrosarcoma/inducido químicamente , PPAR alfa/agonistas , PPAR gamma/agonistas , Adipocitos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Alcanosulfonatos/sangre , Alcanosulfonatos/metabolismo , Análisis de Varianza , Animales , Biomarcadores , Proliferación Celular , Fibrosarcoma/patología , Expresión Génica , Inflamación/inducido químicamente , Masculino , Fenilpropionatos/sangre , Fenilpropionatos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
5.
Toxicol Appl Pharmacol ; 231(1): 1-9, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18455211

RESUMEN

In this work, we investigated a potential mechanism behind the observation of increased aminotransferase levels in a phase I clinical trial using a lipid-lowering drug, the peroxisome proliferator-activated receptor (PPAR) alpha agonist, AZD4619. In healthy volunteers treated with AZD4619, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities were elevated without an increase in other markers for liver injury. These increases in serum aminotransferases have previously been reported in some patients receiving another PPARalpha agonist, fenofibrate. In subsequent in vitro studies, we observed increased expression of ALT1 protein and mRNA in human hepatocytes after treatment with fenofibric acid. The PPAR effect on ALT1 expression was shown to act through a direct transcriptional mechanism involving at least one PPAR response element (PPRE) in the proximal ALT1 promoter, while no effect of fenofibrate and AZD4619 was observed on the ALT2 promoter. Binding of PPARs to the PPRE located at -574 bp from the transcriptional start site was confirmed on both synthetic oligonucleotides and DNA in hepatocytes. These data show that intracellular ALT expression is regulated by PPAR agonists and that this mechanism might contribute to increased ALT activity in serum.


Asunto(s)
Alanina Transaminasa/biosíntesis , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/enzimología , PPAR alfa/agonistas , PPAR alfa/fisiología , Adulto , Alanina Transaminasa/genética , Biomarcadores , Cromatina/genética , Ensayo de Cambio de Movilidad Electroforética , Fenofibrato/análogos & derivados , Fenofibrato/farmacología , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Hipolipemiantes/efectos adversos , Hipolipemiantes/farmacología , Inmunoprecipitación , Luciferasas/genética , Masculino , Mutagénesis/efectos de los fármacos , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , ARN/biosíntesis , ARN/aislamiento & purificación , Elementos de Respuesta/genética , Elementos de Respuesta/fisiología , Transfección
6.
Toxicol Appl Pharmacol ; 207(1): 1-24, 2005 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-16054898

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent hepatotoxin that exerts its toxicity through binding to the aryl hydrocarbon receptor (AhR) and the subsequent induction or repression of gene transcription. In order to further identify novel genes and pathways that may be associated with TCDD-induced hepatotoxicity, we investigated gene changes in rat liver following exposure to single oral doses of TCDD. Male Sprague-Dawley rats were administered single doses of 0.4 microg/kg bw or 40 microg/kg bw TCDD and killed at 6 h, 24 h, or 7 days, for global analyses of gene expression. In general, low-dose TCDD exposure resulted in greater than 2-fold induction of genes coding for a battery of phase I and phase II metabolizing enzymes including CYP1A1, CYP1A2, NADPH quinone oxidoreductase, UGT1A6/7, and metallothionein 1. However, 0.4 microg/kg bw TCDD also altered the expression of Gadd45a and Cyclin D1, suggesting that even low-dose TCDD exposure can alter the expression of genes indicative of cellular stress or DNA damage and associated with cell cycle control. At the high-dose, widespread changes were observed for genes encoding cellular signaling proteins, cellular adhesion, cytoskeletal and membrane transport proteins as well as transcripts coding for lipid, carbohydrate and nitrogen metabolism. In addition, decreased expression of cytochrome P450 7A1, short heterodimer partner (SHP; gene designation nr0b2), farnesyl X receptor (FXR), Ntcp, and Slc21a5 (oatp2) were observed and confirmed by RT-PCR analyses in independent rat liver samples. Altered expression of these genes implies major deregulation of cholesterol metabolism and bile acid synthesis and transport. We suggest that these early and novel changes have the potential to contribute significantly to TCDD induced hepatotoxicity and hypercholesterolemia.


Asunto(s)
Ácidos y Sales Biliares/biosíntesis , Bilis/fisiología , Colesterol/metabolismo , Perfilación de la Expresión Génica , Hígado/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Dibenzodioxinas Policloradas/toxicidad , Animales , Metabolismo de los Hidratos de Carbono , Inactivación Metabólica , Cuerpos Cetónicos/biosíntesis , Lípidos/biosíntesis , Hígado/metabolismo , Masculino , Nitrógeno/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA