Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Dyn ; 236(4): 1044-54, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17326132

RESUMO

The amino acid homocysteine increases in the serum when there is insufficient folic acid or vitamin B(12), or with certain mutations in enzymes important in methionine metabolism. Elevated homocysteine is related to increased risk for cardiovascular and other diseases in adults and elevated maternal homocysteine increases the risk for certain congenital defects, especially those that result from abnormal development of the neural crest and neural tube. Experiments with the avian embryo model have shown that elevated homocysteine perturbs neural crest/neural tube migration in vitro and in vivo. Whereas there have been numerous studies of homocysteine-induced changes in gene expression in adult cells, there is no previous report of a homocysteine-responsive transcriptome in the embryonic neural crest. We treated neural crest cells in vitro with exogenous homocysteine in a protocol that induces significant changes in neural crest cell migration. We used microarray analysis and expression profiling to identify 65 transcripts of genes of known function that were altered by homocysteine. The largest set of effected genes (19) included those with a role in cell migration and adhesion. Other major groups were genes involved in metabolism (13); DNA/RNA interaction (11); cell proliferation/apoptosis (10); and transporter/receptor (6). Although the genes identified in this experiment were consistent with prior observations of the effect of homocysteine upon neural crest cell function, none had been identified previously as response to homocysteine in adult cells.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Homocisteína/farmacologia , Mioblastos Cardíacos/metabolismo , Crista Neural/embriologia , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Embrião de Galinha , Perfilação da Expressão Gênica , Crista Neural/metabolismo
2.
Ann N Y Acad Sci ; 919: 261-77, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11083116

RESUMO

Neural tube defects (NTDs) are among the most common of all human congenital defects, with multifactorial etiologies comprising both environmental and genetic components. Several murine model systems have been developed in an effort to elucidate genetic factors regulating expression of NTDs. Strain-dependent differences in susceptibility to teratogenic insults and altered patterns of gene expression observed within the neuroepithelium of affected embryos support the hypothesis that subtle genetic changes can result in NTDs. Since several affected genes are folate-regulated, transgenic knockout mice lacking a functional folate receptor were developed. Nullizygous embryos died in utero with significant morphological defects, supporting the critical role of folic acid in early embryogenesis. While epidemiological studies have not established an association between polymorphisms in the human folate receptor gene and NTDs, it is known that folate supplementation reduces infant NTD risk. Continued efforts are therefore necessary to reveal the mechanism by which folate works and the nature of the gene(s) responsible for human NTDs.


Assuntos
Poluentes Ambientais/toxicidade , Predisposição Genética para Doença/genética , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Receptores de Superfície Celular , Animais , Proteínas de Transporte/genética , Ciclo Celular/genética , Impressões Digitais de DNA , Modelos Animais de Doenças , Desenvolvimento Embrionário e Fetal/genética , Receptores de Folato com Âncoras de GPI , Ácido Fólico/metabolismo , Ácido Fólico/farmacologia , Idade Gestacional , Substâncias de Crescimento/genética , Substâncias de Crescimento/metabolismo , Humanos , Hipertermia Induzida/efeitos adversos , Camundongos , Camundongos Knockout , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/patologia , Polimorfismo Conformacional de Fita Simples , Ácido Valproico/farmacologia
3.
Ther Drug Monit ; 22(5): 574-81, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11034263

RESUMO

The purpose of this study was to evaluate the stereoselective pharmacokinetics of valnoctamide (VCD) in dogs, rats, and mice; which are the most common animal models for pharmacokinetic, pharmacologic, and toxicologic evaluation; and to compare it with previously published human data. Racemic VCD (mixture of four stereoisomers) was administered intravenously to six mongrel dogs and to rats (five rats per time-point), and intraperitoneally to mice (five mice per time-point). Plasma concentrations of the individual stereoisomers were measured by a stereospecific gas chromatography assay. In dogs, (2S,3R)-VCD had a larger clearance (0.33 L/h x kg) and a larger volume of distribution (0.79 L/kg) than its two diastereomers (0.24-0.25 L/h x kg and 0.65 L/kg, respectively). A tendency toward slightly higher clearance and volume of distribution values for (2S,3R)-VCD was observed in rats and mice as well. Consequently, in all three animal species the half-life (t1/2) of (2S,3R)-VCD was not different from the t1/2 of the other three VCD stereoisomers. The stereoselective pharmacokinetics of VCD as observed in dogs, rats, and mice is in line with the stereoselectivity previously observed in healthy subjects and epileptic patients.


Assuntos
Amidas/farmacocinética , Ansiolíticos/farmacocinética , Modelos Animais , Amidas/sangue , Animais , Ansiolíticos/sangue , Cromatografia Gasosa , Estudos Cross-Over , Cães , Humanos , Masculino , Camundongos , Distribuição Aleatória , Ratos , Estereoisomerismo
4.
Breast Cancer Res Treat ; 60(2): 135-42, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10845276

RESUMO

Microsatellite markers may provide evidence of faulty DNA mismatch repair (MMR) via the detection of microsatellite instability (MSI). The choice of microsatellite markers may impact on the MSI detection rate. In hereditary non-polyposis colon cancer (HNPCC), several informative microsatellite markers have been recommended. Two of these, BAT 25 and BAT 26, are quasi-homozygous, enabling analysis of tumour DNA in the absence of paired normal DNA. Sixty-six breast cancer patients under 45 years of age at diagnosis were examined for MSI at BAT 25 and BAT 26. Tumour DNA was extracted from paraffin-embedded tissue. No MSI was detected at the BAT 25 or BAT 26 loci. An additional five microsatellite markers, known to be informative for HNPCC, were examined for MSI in these patients. Apparently-normal profiles were achieved. A tabulated survey of 306 microsatellite markers used to detect MSI in breast cancer revealed that only 35.5% of markers detected MSI at an average rate of 2.9%. The MSI detection rate at the specific HNPCC markers varied from 0% to 10% in breast cancer, with D175250 and TP53 being the HNPCC markers most suitable for analysis of breast cancer. The size of the microsatellite marker's repeat unit did not impact on MSI detection rates. Compiled data from large studies (n > 100) revealed D115988 as the marker with the highest MSI detection rate. Genomic instability pathways of carcinogenesis, characterised by MMR defects and MSI, appear to play a role in the genesis of some breast cancer types.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , DNA de Neoplasias/genética , Repetições de Microssatélites/genética , Mutação , Adulto , Idade de Início , Neoplasias da Mama/patologia , Neoplasias Colorretais Hereditárias sem Polipose/genética , Feminino , Humanos , Pessoa de Meia-Idade , Sensibilidade e Especificidade
5.
Teratology ; 61(4): 305-13, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10716750

RESUMO

Neural tube defects (NTDs), although prevalent and easily diagnosed, are etiologically heterogeneous, rendering mechanistic interpretation problematic. To date, there is evidence that mammalian neural tube closure (NTC) initiates and fuses intermittently at four discrete locations. Disruption of this process at any of these four sites may lead to a region-specific NTDs, possibly arising through closure site-specific genetic mechanisms. Although recent efforts have focused on elucidating the genetic components of NTDs, a void persists regarding gene identification in closure site-specific neural tissue. To this end, experiments were conducted to identify neural tube closure site-specific genes that might confer regional sensitivity to teratogen-induced NTDs. Using an inbred mouse strain (SWV/Fnn) with a high susceptibility to VPA- induced NTDs that specifically targets and disrupts NTC between the prosencephalon and mesencephalon region (future fore/midbrain; neural tube closure site II), we identified a VPA-sensitive closure site II-specific clone. Sequencing of this clone from an SWV neural tube cDNA library confirmed that it encodes the r1 subunit of the cell cycle enzyme ribonucleotide reductase (RNR). The abundance of rnr-r1 mRNA was significantly increased in response to VPA drug treatment. This upregulated expression was accompanied by a significant decrease in cellular proliferation in the closure site II neural tube region of the embryos, as determined by ELISA cellular proliferation assays performed on BrdU-pulsed neuroepithelial cells in vivo. We hypothesize that rnr-r1 plays a critical role in the development of VPA-induced exencephaly.


Assuntos
Defeitos do Tubo Neural/induzido quimicamente , Ribonucleotídeo Redutases/genética , Teratogênicos , Ácido Valproico/toxicidade , Animais , Anticonvulsivantes/toxicidade , Desenvolvimento Embrionário e Fetal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Biblioteca Gênica , Predisposição Genética para Doença , Camundongos , Camundongos Endogâmicos , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/genética , Gravidez , RNA Mensageiro/genética , Ribonucleosídeo Difosfato Redutase , Transcrição Gênica
6.
Reprod Toxicol ; 14(1): 1-11, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10689198

RESUMO

Although the teratogenicity of valproic acid (VPA) has been well established, the mechanism(s) by which this anticonvulsant drug induces malformations remains controversial. Using the combined molecular techniques of in situ-transcription (IST) and antisense RNA (aRNA) amplification we analyzed VPA-induced alterations in the gene expression for 10 genes within the neural tubes of embryos from two murine strains that have been shown to differ in their susceptibility to VPA-induce neural tube defects (NTD). Pregnant dams from both SWV (susceptible) and LM/Bc (resistant) strains were either treated with saline (control) or VPA (600 mg/kg) on gestational day (GD) 8:12 (day:hour). Neural tubes were isolated from control or VPA exposed embryos at three gestational time points, which represented the beginning (GD 8:18), middle (GD 9:00), and end (GD 9:12) of neural tube closure (NTC) in both of these murine strains. Using univariant statistics we demonstrated that in LM/Bc embryos with NTDs, the expression of bdnf, ngf, and trk, ngf-R were significantly elevated at all three time points, and the cytokine, cntf was significantly decreased at GD 9:00. In contrast, the major gene alterations observed in SWV embryos were a significant increase in tfgalpha and tgfbeta1-3 at GD 9:00. In an effort to better define the more intricate interactions between VPA exposure and the expression of these genes, we analyzed our data using Principal Component Analysis. The results from this analysis demonstrated that embryos from these two stains behaved differently, not only in response to a VPA exposure, but also under control conditions, which may explain the multifactorial nature of NTDs in these mice.


Assuntos
Anticonvulsivantes/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Substâncias de Crescimento/genética , Fatores de Crescimento Neural/genética , Defeitos do Tubo Neural/induzido quimicamente , Teratogênicos/toxicidade , Ácido Valproico/toxicidade , Análise de Variância , Animais , Feminino , Expressão Gênica/efeitos dos fármacos , Substâncias de Crescimento/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos , Fatores de Crescimento Neural/biossíntese , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Gravidez , Transcrição Gênica/efeitos dos fármacos
7.
Pharm Res ; 16(10): 1582-8, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10554101

RESUMO

PURPOSE: The purpose of this study was to evaluate there existed stereoselective effects in the pharmacokinetics, anticonvulsant activity, microsomal epoxide hydrolase (mEH) inhibition, and teratogenicity of the two enantiomers of propylisopropyl acetamide (PID), a CNS-active chiral amide analogue of valproic acid. METHODS: Racemic PID, as well as the individual enantiomers, were intravenously administered to six dogs in order to investigate the stereoselectivity in their pharmacokinetics. Anticonvulsant activity was evaluated in mice (ip) and rats (oral), mEH inhibition studies were performed in human liver microsomes, and teratogenicity was evaluated in an inbred susceptible mice strain. RESULTS: Following intravenous administration to dogs of the individual enantiomers, (R)-PID had significantly lower clearance and longer half-life than (S)-PID, however, the volumes of distribution were similar. In contrast, following intravenous administration of racemic PID, both enantiomers had similar pharmacokinetic parameters. In rats (oral), (R)-PID had a significantly lower ED50 in the maximal electroshock seizure test than (S)-PID; 16 and 25 mg/kg, respectively. PID enantiomers were non-teratogenic and did not demonstrate stereoselective mEH inhibition. CONCLUSIONS: (R)-PID demonstrated better anticonvulsant activity, lower clearance and a longer half-life compared to (S)-PID. When racemic PID was administered, the clearance of (S)-PID was significantly reduced, reflecting an enantiomer-enantiomer interaction.


Assuntos
Alilisopropilacetamida/análogos & derivados , Anticonvulsivantes/farmacologia , Anticonvulsivantes/farmacocinética , Ácido Valproico/análogos & derivados , Alilisopropilacetamida/farmacocinética , Alilisopropilacetamida/farmacologia , Alilisopropilacetamida/toxicidade , Animais , Anticonvulsivantes/toxicidade , Proteínas Sanguíneas/metabolismo , Cromatografia Gasosa , Cães , Epóxido Hidrolases/metabolismo , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Gravidez , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Teratogênicos/toxicidade , Ácido Valproico/farmacocinética , Ácido Valproico/farmacologia
8.
Nat Genet ; 23(2): 228-32, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508523

RESUMO

Periconceptional folic acid supplementation reduces the occurrence of several human congenital malformations, including craniofacial, heart and neural tube defects. Although the underlying mechanism is unknown, there may be a maternal-to-fetal folate-transport defect or an inherent fetal biochemical disorder that is neutralized by supplementation. Previous experiments have identified a folate-binding protein (Folbp1) that functions as a membrane receptor to mediate the high-affinity internalization and delivery of folate to the cytoplasm of the cell. In vitro, this receptor facilitates the accumulation of cellular folate a thousand-fold relative to the media, suggesting that it may be essential in cytoplasmic folate delivery in vivo. The importance of an adequate intracellular folate pool for normal embryogenesis has long been recognized in humans and experimental animals. To determine whether Folbp1 is involved in maternal-to-fetal folate transport, we inactivated Folbp1 in mice. We also produced mice lacking Folbp2, another member of the folate receptor family that is GPI anchored but binds folate poorly. Folbp2-/- embryos developed normally, but Folbp1-/- embryos had severe morphogenetic abnormalities and died in utero by embryonic day (E) 10. Supplementing pregnant Folbp1+/- dams with folinic acid reversed this phenotype in nullizygous pups. Our results suggest that Folbp1 has a critical role in folate homeostasis during development, and that functional defects in the human homologue (FOLR1) of Folbp1 may contribute to similar defects in humans.


Assuntos
Proteínas de Transporte/genética , Desenvolvimento Embrionário e Fetal/genética , Receptores de Superfície Celular , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Feminino , Morte Fetal/genética , Receptor 1 de Folato , Receptores de Folato com Âncoras de GPI , Ácido Fólico/sangue , Genótipo , Homocisteína/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Gravidez
9.
Reprod Toxicol ; 13(2): 85-91, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10213515

RESUMO

PURPOSE: Previous studies have suggested that polytherapy by design may aid in the management of human pregnancies complicated by epilepsy. However, mechanistic parallels must be drawn between the models of teratogenesis and human pregnancies, and doses of the second agent given to minimize side-effects must be justified. This study sought to determine the lowest dosage of stiripentol (STP) protective against phenytoin-induced teratogenesis in a mouse model, and to determine mechanistically if inhibition of oxidative metabolism by STP in vitro decreased production of reactive phenytoin (PHT) metabolites. METHODS: Pregnant SWV mice were assigned to control or treatment groups of STP alone, PHT alone, or PHT with ascending doses of STP coadministration. Treatments continued from Day 6 to Day 18 of gestation when fetuses were examined for developmental anomalies. [14C]PHT was incubated in mouse liver microsomes with and without NADPH and in the presence or absence of STP or piperonyl butoxide. Covalent binding of [14C] was measured. RESULTS: There were no dose-related differences in the frequency of fetal malformations per litter among groups treated with STP alone. However, STP (all doses) reduced the frequency of PHT-induced malformations. Covalent binding of [14C]PHT was NADPH-dependent and was inhibited by either piperonyl butoxide or STP. CONCLUSIONS: The beneficial effects of STP occurred at concentrations below the therapeutic range for its anticonvulsant effects. These results support the concept of polytherapy by design to reduce the risk of teratogenesis associated with PHT.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Anormalidades Induzidas por Medicamentos/prevenção & controle , Anticonvulsivantes/toxicidade , Anticonvulsivantes/uso terapêutico , Dioxolanos/uso terapêutico , Fenitoína/toxicidade , Administração Oral , Animais , Anticonvulsivantes/sangue , Radioisótopos de Carbono , Dioxolanos/sangue , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Camundongos , Camundongos Endogâmicos , NADP/farmacologia , Especificidade de Órgãos , Fenitoína/sangue , Gravidez
10.
Teratology ; 59(1): 23-34, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9988880

RESUMO

In utero exposure to the anticonvulsant drug phenytoin has been shown to alter normal embryonic development, leading to a pattern of dysmorphogenesis known as the Fetal Hydantoin Syndrome. This embryopathy is characterized by growth retardation, microcephaly, mental deficiency, and craniofacial malformations, although the precise mechanism(s) by which phenytoin alters normal developmental pathways remains unknown. To better understand the molecular events involved in the pathogenesis of phenytoin-induced congenital defects, alterations in gene expression were examined during critical periods of craniofacial development. Pregnant SWV mice were administered phenytoin (60 mg/kg/day) from gestational day 6.5 until they were sacrificed at selected developmental time points. Tissue from the craniofacial region of control and exposed embryos was isolated, and samples were subjected to in situ transcription, antisense RNA amplification, and hybridization on reverse Northern blots to quantitatively assess expression of 36 candidate genes. Chronic phenytoin exposure significantly altered expression of several genes at distinct times during morphogenesis. Results of these studies show that expression of the retinoic acid receptors (RAR) alpha, beta, and gamma were significantly increased by phenytoin exposure. Elevations in gene expression of laminin beta 1, and the growth factors IGF-2, TGF alpha, and TGF beta 1, were also demonstrated in the craniofacial region of phenytoin-exposed embryos. As several of these genes are transcriptionally regulated by retinoic-acid-responsive elements in their promoter regions, phenytoin-induced alterations in expression of the RAR isoforms may have severe downstream consequences in the regulation of events necessary for normal craniofacial development. Such alterations occurring coordinately at critical times during craniofacial development may account for the dysmorphogenesis often associated with phenytoin exposure.


Assuntos
Anticonvulsivantes/toxicidade , Expressão Gênica/efeitos dos fármacos , Fenitoína/toxicidade , Anormalidades Induzidas por Medicamentos/genética , Animais , DNA Complementar , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Face/embriologia , Feminino , Camundongos , Camundongos Endogâmicos , Morfogênese , Gravidez , Receptores do Ácido Retinoico/genética , Crânio/embriologia
11.
Mol Genet Metab ; 66(1): 31-9, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9973545

RESUMO

Expression patterns of mRNAs coding for the murine folate binding proteins one and two (FBP1 and FBP2) were determined by ribonuclease protection assay (RPA) in highly inbred SWV/Fnn mouse embryos. Tissue samples for RPA were collected from the anterior neural tube throughout the period of embryonic development, as well as from maternal- and fetal-derived term placenta. The peak in expression of FBP1 occurred in term placental tissue compared to neural tissue from any time point. This relative increase in FBP1 expression occurred in placental tissue of embryonic, as opposed to maternal, origin. The expression of FBP2 did not differ statistically between any timepoints or tissues examined. Expression of both FBP1 and FBP2 was slightly elevated throughout the period of neural tube closure (Gestational Days 8 through 10), although not significantly. These data fit the anticipated expression patterns of the homologues of human folate receptors alpha and beta, thus helping to resolve some of the confusion secondary to the nomenclature associated with this gene family. Furthermore, the expression of these two genes in the neural tube closure stage of embryological development supports their involvement in regulatory events related to normal neural tube morphogenesis.


Assuntos
Proteínas de Transporte/genética , Desenvolvimento Embrionário e Fetal/genética , Receptores de Superfície Celular , Análise de Variância , Animais , Embrião de Mamíferos/metabolismo , Feminino , Receptores de Folato com Âncoras de GPI , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Placenta/metabolismo , Gravidez , RNA Mensageiro/análise , RNA Mensageiro/genética
12.
Teratology ; 57(1): 17-29, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9516748

RESUMO

The murine mutant Splotch (Sp) is a well-established model for studying neural tube closure defects. In the current investigation, the progression through neural tube closure (NTC) as well as the expression patterns of 12 developmentally regulated genes were examined in the neural tissue of wildtype (+/+), Splotch heterozygous (Sp/+), and Splotch homozygous (Sp/Sp) embryos during neurulation. The overall growth of the embryos, as measured by the number of somite pairs, did not differ significantly between the three genotypes at any of the collection time-points. There was, however, a significant delay in the progression through NTC for both the Sp/+ and Sp/Sp embryos. A univariate analysis on the expression of the 12 candidate genes (bcl-2, FBP-2, Hmx-2, Msx-3, N-cam, N-cad, noggin, p53, Pax-3, Shh, Wee-1, wnt-1) revealed that although 11 were statistically altered, across time or by genotype, there were no significant interactions between gestation age and genotype for any of these genes during NTC. However, a multivariate statistical analysis on the simultaneous expression of these genes revealed interactions at both gestation day (GD) 8:12 (day:hour) and 9:00 among Pax-3, N-cam, N-cad, bcl-2, p53, and Wee-1 that could potentially explain the aberrant NTC. The data from these studies suggest that a disruption in the genes that govern the cell cycle or extracellular matrices of the developing neural tube might play a critical role in the occurrence of the NTDs observed in Splotch embryos.


Assuntos
Sistema Nervoso Central/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos Mutantes Neurológicos/embriologia , Defeitos do Tubo Neural/genética , Animais , Moléculas de Adesão Celular/genética , Análise Mutacional de DNA , Modelos Animais de Doenças , Genes/genética , Genes/fisiologia , Idade Gestacional , Heterozigoto , Proteínas de Homeodomínio/genética , Homozigoto , Camundongos , Análise Multivariada , Defeitos do Tubo Neural/embriologia , Proteínas Nucleares/genética , Somitos
13.
Drug Metab Dispos ; 25(8): 953-62, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9280403

RESUMO

The disposition of carbamazepine (CBZ) was investigated in the SWV mouse. A 14C-CBZ dose was administered to CBZ pretreated mice, and the distribution of radiolabeled material was determined. Twenty-four hours after the 14C-CBZ dose, 92.5% of the dose was accounted for in urine (56%), in the visera and carcass (22%), in feces (11%), and expired as 14CO2 (2%). CBZ metabolites present in hydrolyzed urine were also identified using a combination of spectroscopic techniques. CBZ, CBZ-10,11-epoxide (CBZE), 2- and 3-hydroxy-CBZ, methylsulfonyl-CBZ, and glucuronides of CBZ and CBZE accounted for 64% of total urinary radioactivity (0-24 hr) in CBZ pretreated mice. Minor metabolites of CBZ included novel cysteine and N-acetylcysteine conjugates of CBZ, as well as a methylsulfonyl conjugate of CBZE not previously reported. The urinary excretion of these thioether conjugates was increased in CBZ/phenobarbital pretreated mice and decreased in CBZ/stiripentol pretreated mice in comparison with CBZ-only treated mice. Preliminary studies of the effects of phenobarbital and stiripentol on the urinary abundance of these metabolites are consistent with the modulation of teratogenicity in the SWV mouse by the same pretreatments. These data suggest the formation of thioether metabolites of CBZ may be related to CBZ teratogenicity in the SWV mouse.


Assuntos
Carbamazepina/metabolismo , Teratogênicos/metabolismo , Animais , Carbamazepina/toxicidade , Radioisótopos de Carbono , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos , Teratogênicos/toxicidade
14.
Am J Med Genet ; 70(3): 303-11, 1997 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-9188671

RESUMO

The molecular basis for the well-established hierarchy of susceptibility to valproic acid-induced neural tube defects in inbred mouse strains was examined using in situ transcription and anti-sense RNA amplification methodologies with both univariate and multivariate analyses of the resulting gene expression data. The highly sensitive SWV strain demonstrated a significant reduction in the expression of the folate binding protein (FBP-1) following the teratogenic insult at gestational day 8:18, while the more resistant LM/Bc embryos were up-regulating this gene in response to valproic acid treatment. More importantly, at all 3 gestational timepoints spanning the period of murine neural tube closure examined in this study, the LM/Bc embryos had significantly higher MTHFR (5,10-methylenetetrahydrofolate reductase) gene expression levels compared to the SWV embryos. As this folate pathway enzyme is important in homocysteine and methionine metabolism, it suggests that the SWV embryos may be hypomethylated, and essential gene expression during critical periods of neural tube closure is compromised by the teratogenic exposure to valproic acid. This study represents the first evidence of a strain difference in transcriptional activity in response to a teratogenic exposure that might be causally related to the development of the teratogen-induced congenital malformations.


Assuntos
Defeitos do Tubo Neural/induzido quimicamente , Teratogênicos/toxicidade , Ácido Valproico/toxicidade , 5,10-Metilenotetra-Hidrofolato Redutase (FADH2) , Animais , Interpretação Estatística de Dados , Camundongos , Modelos Biológicos , Defeitos do Tubo Neural/enzimologia , Oxirredutases/genética , Polimorfismo Genético , Especificidade da Espécie
15.
Epilepsia ; 38(4): 415-23, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9118846

RESUMO

PURPOSE: We wished to determine whether chronic phenytoin (PHT) exposure could impair neural development and if any morphological alterations could be linked to changes in gene expression. METHODS: Pregnant SWV mice were chronically administered PHT 40 mg/kg/day from gestational day (GD) 0:12 (day:h) until they were killed at various timepoints throughout neural tube closure (NTC). At each timepoint, embryos from both treated and control dams were collected and scored for their progression through NTC. The neural tubes were then isolated and subjected to in situ transcription (IST) and antisense RNA amplification procedures. Using these techniques, we examined the expression of 10 genes: N-cadherin (Ncad), collagen type IV (col-IV), bcl-2, c-jun, PAX-3, collular retinol binding protein-2 (CRBP-2), retinoic acid receptor alpha (RAR alpha), transforming growth factor(beta2) (TGF(beta2)), wee-1, and EMX-2. RESULTS: Chronic PHT exposure not only caused a delay in NTC whereby exposed embryos lagged behind the controls at each collection timepoint, but also significantly altered the expression of specific genes at distinct times during NTC. Early in NTC, PHT induced a significant reduction in the expression of N-cad, col-IV, and c-jun in exposed embryos as compared with controls. In contrast, during the midstages of NTC, the only significant molecular alterations observed in the PHT-exposed embryos was the continued decreased expression of col-IV and an increase in CRBP-2 expression. Finally, in the latter stages of NTC, PHT caused a significant reduction in the expression of bcl-2, RAR alpha, TGF(beta2), EMX-2, and PAX-3. CONCLUSIONS: These results show that although the effects of PHT are morphologically subtle, causing a delay in the development of the neural tube, this delay is accompanied by alterations in critical genes at crucial times of neural development that may account for the observed neurological deficits often associated with PHT exposure.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Proteínas de Ciclo Celular , Defeitos do Tubo Neural/induzido quimicamente , Proteínas Nucleares , Fenitoína/toxicidade , Fatores de Transcrição , Anormalidades Induzidas por Medicamentos/genética , Animais , Caderinas/biossíntese , Caderinas/genética , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/embriologia , Colágeno/biossíntese , Colágeno/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Feminino , Amplificação de Genes , Expressão Gênica/efeitos dos fármacos , Genes jun/efeitos dos fármacos , Genes jun/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Defeitos do Tubo Neural/genética , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Gravidez , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Receptores do Ácido Retinoico/biossíntese , Receptores do Ácido Retinoico/genética , Proteínas de Ligação ao Retinol/biossíntese , Proteínas de Ligação ao Retinol/genética , Proteínas Celulares de Ligação ao Retinol
16.
Biochem Mol Med ; 60(2): 81-91, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9169087

RESUMO

The molecular techniques of in situ transcription and antisense RNA amplification (IST/aRNA) have allowed for the monitoring of coordinate changes in the expression of multiple genes simultaneously. However, the analysis of their concurrent behavior during murine embryogenesis has been problematic. Studies involving the investigation of temporal and spatial gene expression during embryogenesis have focused solely on the analysis of isolated, single gene events. Such an approach has failed to provide an integrative picture of genetic control over the varied and complicated cellular processes governing embryogenesis. In order to interpret the enormous amount of gene expression data generated by these procedures, we have attempted to develop an analytical framework by employing the statistical concepts of principal components analysis (PCA). For the current study, we performed IST/aRNA on neural tubes dissected from the highly inbred LM/Bc murine strain collected during four gestational time periods. A subset of these genes, representing a partial signaling pathway in the developing neuroepithelium, was then subjected to PCA. Here, we report that PCA highlighted the transcriptional interplay among the genes p53, wee-1, Tgf beta-2, and bcl-2 such that the combined reciprocal regulation of their gene products is suggestive of a predominant proliferative state for the developing neuroepithelium. The application of PCA to the gene expression data has elucidated previously unknown interrelationships among cell cycle genes, growth, and transcription factors on a transcriptional level during critical stages of neurulation. The information gleaned from this analysis, while not definitive, suggests distinct hypotheses to guide future research.


Assuntos
Proteínas de Ciclo Celular , Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas Genéticas , Proteínas Nucleares , Análise de Variância , Animais , Ciclo Celular/genética , DNA Complementar/genética , Feminino , Genes bcl-2/genética , Genes fos , Genes p53 , Idade Gestacional , Camundongos , Camundongos Endogâmicos , Sistema Nervoso/embriologia , Gravidez , Proteínas Tirosina Quinases/genética , RNA Antissenso/genética , Transcrição Gênica , Fator de Crescimento Transformador beta/genética
17.
Dev Genet ; 21(3): 212-22, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9397537

RESUMO

Neural tube defects (NTDs) are among the most common congenital malformations, affecting approximately 1 per 1,000 liveborn infants in the United States [Nakano, 1973; Richards et al., 1972]. Maternal exposure to hyperthermia, either through recreational sources or due to an infectious agent, is thought to account for approximately 10% of observed NTD cases. The specific genes conferring susceptibility or resistance to hyperthermia-induced NTDs have not been identified. This study used differential display-polymerase chain reaction (DD-PCR) to characterize alterations in gene expression in the anterior embryonic neural tube of two highly inbred murine strains (SWV/Fnn, LM/Bc/Fnn) known to differ in their genetically determined susceptibility to heat-induced NTDs. Herein, we report the neural tube-specific differential expression of the growth arrest specific (gas 5) gene in the highly susceptible SWV/Fnn strain during neural tube closure (NTC). Although the expression of gas 5 did not appear to be altered by the teratogenic heat treatment, its spatial and strain-specific pattern of expression makes it an excellent candidate gene responsible for the observed genetic differences in NTD susceptibility between these two inbred murine strains.


Assuntos
Febre/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Camundongos Endogâmicos/genética , Defeitos do Tubo Neural/genética , RNA Nucleolar Pequeno , Animais , Sequência de Bases , Feminino , Predisposição Genética para Doença , Camundongos , Dados de Sequência Molecular , Gravidez , Alinhamento de Sequência
18.
J Pharmacol Exp Ther ; 279(3): 1237-42, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8968346

RESUMO

The mechanism of carbamazepine (CBZ)-related teratogenicity was investigated in the SWV mouse by contrasting the effects of CBZ-10, 11-epoxide (CBZE) and oxcarbazepine (OXC) treatments. Dietary CBZE administration was initiated 2 weeks before mating and continued through day 18 of gestation. OXC was administered to pregnant dams by gavage on day 6 of gestation and continued through day 18 of gestation. Maternal plasma concentrations of CBZE ranged from 1.4 to 17.7 micrograms/ml and OXC ranged from 6.1 to 15.9 micrograms/ml. In comparison, clinical plasma concentrations of CBZE ranged from 1 to 2 micrograms/ml and OXC plasma concentrations were 1 microgram/ml or less. The incidence of malformation were 14%, 27% and 26% after daily CBZE doses of 300, 600 and 1000 mg/kg, respectively, compared with a 6% incidence in no-drug control mice, P < .05. The incidence of malformation was 8% after exposure at the highest tolerable dose of OXC (1100 mg/kg/day), compared with a 5% incidence in no-drug controls, P > .05. Phenobarbital cotreatment (45 mg/kg/day) with OXC (1100 mg/kg/day) did not lead to changes in the incidence of malformation when compared with OXC (1100 mg/kg/day) dosed alone. These data are consistent with a teratogenic CBZ metabolite, possibly CBZE, or with oxidation of CBZE or CBZ at positions on the aromatic ring leading to the formation of reactive intermediates such as arene oxides or quinones.


Assuntos
Anticonvulsivantes/toxicidade , Carbamazepina/análogos & derivados , Teratogênicos/toxicidade , Animais , Carbamazepina/toxicidade , Feminino , Camundongos , Camundongos Endogâmicos , Oxcarbazepina , Gravidez
19.
Teratology ; 54(6): 284-97, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9098922

RESUMO

The teratogenic potential of valproic acid has been well established both in experimental models and in human clinical studies. As with all human teratogens, there are genetically determined differences in individual susceptibility to the induction of congenital defects. Using a mouse model of valproate-induced neural tube defects, a study was undertaken to examine differential changes in gene expression for selected transcription factor (Pax-3, Emx-1, Emx-2, c-fos, c-jun, creb) and cell cycle checkpoint genes (bcl-2, p53, wee-1) during neural tube closure. In general, exposure to teratogenic concentrations of valproic acid elicited GD 9:12 control levels of transcription factor mRNA expression in GD 9:0 embryos of both strains. This accelerated developmental profile is marked by significant elevation of Emx-1, Emx-2, c-fos, c-jun, and creb expression. There was also a significant over expression of the cell cycle genes p53 and bcl-2 in the LM/Bc embryos in response to the teratogenic insult. Examination of the ratio of expression of these genes clearly favored bcl-2, which supports the hypothesis that altered neuroepithelial cell proliferation rates, rather than increased apoptosis, is the underlying mechanism by which valproic acid alters normal neural tube morphogenesis. An investigation into interactive effects of these genes on the molecular profile of GD 9:0 embryos further validated this observation. That is, the overall proliferative state among the control embryos was prematurely modified into a more differentiated state following teratogenic insult. These results suggest that alterations in the expression of multiple genes are most likely responsible for valproic acid-induced neural tube defects.


Assuntos
Expressão Gênica/efeitos dos fármacos , Sistema Nervoso/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Teratogênicos/toxicidade , Ácido Valproico/toxicidade , Animais , Camundongos , Camundongos Endogâmicos , Modelos Biológicos , Análise Multivariada , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo
20.
Reprod Toxicol ; 10(6): 447-54, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8946558

RESUMO

The potential of arsenic to cause neural tube defects (NTD) in the human population remains a topic of controversy. While clearly toxic, the lack of well-defined human epidemiologic studies on this subject has made it difficult to fully understand the effects arsenic may have on the developing human neural tube. In the absence of good clinical data, we have tried to develop a murine model where hypotheses about the reproductive toxicity of arsenate can be tested. For these studies a murine strain (LM/Bc) that has proven to be susceptible to arsenic-induced NTD was use. Because cellular proliferation is vital for normal neural tube closure (NTC) to occur, in the present study we investigated whether an acute arsenate treatment could alter the expression of several cell cycle genes during murine neurulation. Pregnant LM/Bc dams were injected intraperitoneally on gestation day (GD) 7:12 (day:hour) and 8:12 with 40 mg/kg of arsenate, a treatment that causes exencephaly in 90 to 100% of the exposed fetuses. Neural tubes were then isolated from both control and arsenic treated embryos at GD 9:00, 9:12, 10:00, and 10:12, which encompasses all the stages of neurulation for this murine strain. Using the molecular techniques of in situ transcription and antisense RNA amplification (RT/aRNA) the expression pattern for bc1-2, p53, wee-1, and wnt-1 was analyzed at each of these time points. In the neural tubes isolated from control embryos, the expression of all four genes was significantly altered as neurulation progressed, demonstrating their developmental regulation. Following arsenate treatment, however, there was a significant upregulation in the expression of bc1-2 and p53 at gestational day 9:0, compared to their control values. The heightened expression of both of these genes suggests that arsenic inhibits cell proliferation, rather than inducing apoptosis, which delayed NTC and ultimately led to the neural tube defects observed in exposed embryos.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Arseniatos/toxicidade , Genes cdc/efeitos dos fármacos , Crista Neural/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Teratogênicos/toxicidade , Animais , Feminino , Expressão Gênica/efeitos dos fármacos , Camundongos , Crista Neural/embriologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...