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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 16(3): 608-15, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703271

RESUMO

Renal tubular reabsorption of filtered folate is essential for the conservation and normal homeostasis of this important vitamin. Different molecular mechanisms have been implicated in epithelial folate transport, including folate receptors. Defective expression or antibody inactivation of these is associated with embryonic defects also correlated with low folate intake; however, their contribution to renal tubular folate reabsorption has not been established. With the use of targeted inactivation of the folate binding protein 1 (folbp1) and folate binding protein 2 (folbp2) genes in mice, the role of folate receptors in renal epithelial folate reabsorption was evaluated during low and normal folate intake. Inactivation of folbp1 was associated with (1) loss of (3)H-folic acid binding to crude kidney membranes, (2) increase in renal folate clearance, and (3) increase in urinary excretion and decrease in renal uptake of injected (3)H-methyltetrahydrofolate. No changes in renal folate handling were observed as a result of folbp2 inactivation. Thus, folbp1 is essential for normal renal tubular folate reabsorption, preventing excessive urinary folate loss. Folbp1 is heavily expressed in choroid plexus, yolk sac, and placenta, supporting a role of folbp1 in folate transport in other tissues. The greatest significance of folbp1 for renal folate uptake was observed at conditions of low folate intake, providing a possible explanation for the ability of folate supplementation to prevent developmental defects associated with folbp1 inactivation.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/sangue , Ácido Fólico/urina , Túbulos Renais/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Animais , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Receptores de Folato com Âncoras de GPI , Ácido Fólico/farmacocinética , Expressão Gênica , Túbulos Renais/citologia , Masculino , Camundongos , Camundongos Knockout , Trítio
2.
Dev Dyn ; 231(1): 221-31, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15305303

RESUMO

Inactivation of folate binding protein-1 (Folbp1) adversely impacts murine embryonic development, as nullizygous embryos (Folbp1(-/-)) die in utero. Administration of folinic acid (N5-formyl-tetrahydrofolate) to Folbp1-deficient dams before and throughout gestation rescues the majority of embryos from premature death; however, a portion of surviving embryos develop structural malformations, including neural tube defects. We examined whether maternal supplementation with L-N5-methyl-tetrahydrofolate (L-5M-THF) has superior protective effects on embryonic development of Folbp1(-/-) fetuses compared with L-N5-formyl-tetrahydrofolate (L-5F-THF). We also examined the critical period during gestation when folate supplementation is most beneficial to the developing Folbp1(-/-) embryos. Folbp1(-/-) pups presented with a range of malformations involving the neural tube, craniofacies, eyes, and abdominal wall. The frequencies of these malformations decreased with increasing folate dose, regardless of the form used. There was no additional benefit provided by L-5M-THF compared with L-5F-THF. Despite rescuing the phenotype in Folbp1(-/-) embryos, no significant elevation of Folbp1(-/-) maternal folate levels was observed with supplementation.


Assuntos
Proteínas de Transporte/genética , Embrião de Mamíferos/anormalidades , Leucovorina/farmacologia , Fenótipo , Receptores de Superfície Celular/genética , Animais , Proteínas de Transporte/metabolismo , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Feminino , Receptores de Folato com Âncoras de GPI , Leucovorina/farmacocinética , Camundongos , Camundongos Knockout , Gravidez , Receptores de Superfície Celular/metabolismo , Taxa de Sobrevida
3.
Neurochem Res ; 29(6): 1105-12, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15176467

RESUMO

Inactivation of the murine folate binding protein-1 (Folbp1) has been shown to play a vital role in embryonic development. Nullizygous embryos (Folbp1-/-) have significant malformations of the neural tube, craniofacies, and conotruncus, and invariably die in utero by gestational day (E) 10. Administration of 25 mg x kg(-1) x day(-1) folinic acid to dams prior to and throughout gestation rescues the majority of embryos from premature death; however, a portion of surviving embryos develops neural tube defects. Using antisense RNA amplification and cDNA microarrays, we examined the expression of approximately 5700 genes in the anterior neural tube of gestational day 9 Folbp1-/- embryos that were supplemented with folinic acid. Genes that appear to be folate regulated include transcription factors, G-proteins, growth factors, methyltransferases, and those that are related to cell proliferation. The potential impact of such changes during neural tube closure is considered in light of the phenotype of Folbp1-/- embryos.


Assuntos
Proteínas de Transporte/genética , Ácido Fólico/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Sistema Nervoso/embriologia , Receptores de Superfície Celular/genética , Animais , Desenvolvimento Embrionário e Fetal/genética , Receptores de Folato com Âncoras de GPI , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Heterozigoto , Camundongos , Camundongos Knockout , Família Multigênica , Hibridização de Ácido Nucleico
4.
Br J Pharmacol ; 139(4): 755-64, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12812999

RESUMO

1 The purpose of this study was to synthesize novel valproyltaurine (VTA) derivatives including valproyltaurinamide (VTD), N-methyl-valproyltaurinamide (M-VTD), N,N-dimethyl-valproyltaurinamide (DM-VTD) and N-isopropyl-valproyltaurinamide (I-VTD) and evaluate their structure-pharmacokinetic-pharmacodynamic relationships with respect to anticonvulsant activity and teratogenic potential. However, their hepatotoxic potential could not be evaluated. The metabolism and pharmacokinetics of these derivatives in mice were also studied. 2 VTA lacked anticonvulsant activity, but VTD, DM-VTD and I-VTD possessed anticonvulsant activity in the Frings audiogenic seizure susceptible mice (ED(50) values of 52, 134 and 126 mg kg(-1), respectively). 3 VTA did not have any adverse effect on the reproductive outcome in the Swiss Vancouver/Fnn mice following a single i.p. injection of 600 mg kg(-1) on gestational day (GD) 8.5. VTD (600 mg kg(-1) at GD 8.5) produced an increase in embryolethality, but unlike valproic acid, it did not induce congenital malformations. DM-VTD and I-VTD (600 mg kg(-1) at GD 8.5) produced a significant increase in the incidence of gross malformations. The incidence of birth defects increased when the length of the alkyl substituent or the degree of N-alkylation increased. 4 In mice, N-alkylated VTDs underwent metabolic N-dealkylation to VTD. DM-VTD was first biotransformed to M-VTD and subsequently to VTD. I-VTD's fraction metabolized to VTD was 29%. The observed metabolic pathways suggest that active metabolites may contribute to the anticonvulsant activity of the N-alkylated VTDs and reactive intermediates may be formed during their metabolism. In mice, VTD had five to 10 times lower clearance (CL), and three times longer half-life than I-VTD and DM-VTD, making it a more attractive compound than DM-VTD and I-VTD for further development. VTD's extent of brain penetration was only half that observed for the N-alkylated taurinamides suggesting that it has a higher intrinsic activity that DM-VTD and I-VTD. 5 In conclusion, from this series of compounds, although VTD caused embryolethality, this compound emerged as the most promising new antiepileptic drug, having a preclinical spectrum characterized by the highest anticonvulsant potential, lowest potential for teratogenicity and favorable pharmacokinetics.


Assuntos
Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/farmacocinética , Teratogênicos/toxicidade , Ácido Valproico/farmacocinética , Animais , Anticonvulsivantes/síntese química , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Injeções Intraperitoneais , Camundongos , Camundongos Mutantes , Estrutura Molecular , Convulsões/tratamento farmacológico , Teratogênicos/farmacocinética , Ácido Valproico/síntese química , Ácido Valproico/uso terapêutico
5.
Birth Defects Res A Clin Mol Teratol ; 67(12): 974-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14745917

RESUMO

BACKGROUND: Folate is an important B vitamin that is transported into cells by way of folate-binding proteins and transporters. Folate-binding protein-2 nullizygous (Folbp2(-/-)) mice develop normally; however, we have found them to be more susceptible to the teratogenic effects of arsenate exposure than wild-type control mice. METHODS: In the current study, we wanted to extend our findings and test the hypothesis that Folbp2(-/-) mice are more susceptible to the teratogenic effects of valproic acid (VPA), a commonly used antiepileptic drug that is known to induce neural tube defects (NTDs) in both humans and laboratory animals. RESULTS: Folbp2(-/-) mice had higher VPA-induced frequencies of embryonic lethality and exencephaly than did the wild-type control mice during folate supplementation and a control diet, respectively. All other differences in response between the two genotypes were short of reaching statistical significance. Folate supplementation of wild-type, but not Folbp2(-/-) dams reduced embryonic lethality of VPA-treated wild-type embryos compared to the folate-deficient diet. CONCLUSIONS: Unlike our previous findings with arsenate, enhanced susceptibility of Folbp2(-/-) mice to in utero VPA exposure was demonstrated in some dietary folate regimens. Thus, our data indicate a relatively frail relationship between Folbp2 and VPA-induced NTDs.


Assuntos
Anormalidades Induzidas por Medicamentos , Anticonvulsivantes/toxicidade , Proteínas de Transporte/genética , Defeitos do Tubo Neural/induzido quimicamente , Receptores de Superfície Celular/genética , Ácido Valproico/toxicidade , Animais , Anticonvulsivantes/administração & dosagem , Proteínas de Transporte/metabolismo , Dieta , Modelos Animais de Doenças , Perda do Embrião/induzido quimicamente , Perda do Embrião/patologia , Perda do Embrião/prevenção & controle , Feminino , Receptores de Folato com Âncoras de GPI , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Ácido Fólico/uso terapêutico , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Defeitos do Tubo Neural/patologia , Defeitos do Tubo Neural/prevenção & controle , Gravidez/sangue , Receptores de Superfície Celular/metabolismo , Ácido Valproico/administração & dosagem
6.
J Nutr ; 132(8 Suppl): 2457S-2461S, 2002 08.
Artigo em Inglês | MEDLINE | ID: mdl-12163711

RESUMO

Periconceptional folic acid supplementation has been shown to prevent up to 70% of neural tube and other birth defects in humans; however, the mechanism is still unknown. In this study, we tested whether defective intracellular folate transport, as achieved by inactivation of the murine folate-binding protein 1 (Folbp1), affects global DNA methylation in the liver and brain from gestational day (GD) 15 embryos. Complete Folbp1 inactivation is embryolethal but can be reversed by maternal folinic acid (FA) supplementation, and thus we also tested the effect of FA supplementation on DNA methylation in Folbp1 fetuses. Overall, the extent of global DNA methylation seems to be similar across all genotypes in unsupplemented control Folbp1 mice; however, explicit conclusions regarding Folbp1(-/-) fetuses were not possible because only a single living unsupplemented fetus was viable at GD 15. FA supplementation induced global DNA hypomethylation across all genotypes. FA-induced hypomethylation is most likely due to its ability to inhibit the enzyme glycine hydroxymethyltransferase, thereby inhibiting the homocysteine remethylation cycle necessary to regenerate S-adenosylmethionine, the methyl donor for DNA methyltransferases. Our hypothesis was that due to defective folate transport in Folbp1(-/-) embryos and fetuses, DNA would be hypomethylated, thereby altering the temporal expression of critical genes necessary for normal embryonic development. However, these results suggest that an extended examination of changes in DNA methylation prior to GD 15 is required to unequivocally prove or disprove the hypothesis.


Assuntos
Proteínas de Transporte/fisiologia , Metilação de DNA , Ácido Fólico/administração & dosagem , Receptores de Superfície Celular , Animais , Proteínas de Transporte/genética , Feminino , Receptores de Folato com Âncoras de GPI , Camundongos , Camundongos Knockout , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA