Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
World Neurosurg ; 134: e822-e825, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31715407

RESUMO

OBJECTIVE: Erythrosine (E127), a synthetic food dye containing iodine and sodium, has often been used inside packaged foods and beverages in Turkey and many other countries. We evaluated the effects of erythrosine on neural tube development in early-stage chicken embryos. METHODS: The study included 4 groups, with a total of 80 embryos: a control group, a normal saline group, a half-dose group, and a high-dose group. After 30 hours of incubation, saline and erythrosine solution was injected under the embryonic discs. At the end of 72 hours, the embryos were excised and evaluated macroscopically and histopathologically. RESULTS: Neural tube defects were detected in the erythrosine-administered groups with statistically significant differences. In contrast, the embryos in the control and saline groups displayed normal development. CONCLUSIONS: Erythrosine increased the risk of neural tube defects in early-stage chicken embryos, even at half of the approved dose.


Assuntos
Eritrosina/farmacologia , Corantes Fluorescentes/farmacologia , Defeitos do Tubo Neural/embriologia , Tubo Neural/efeitos dos fármacos , Anormalidades Induzidas por Medicamentos/embriologia , Anormalidades Induzidas por Medicamentos/etiologia , Animais , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/embriologia , Defeitos do Tubo Neural/induzido quimicamente
2.
Folia Morphol (Warsz) ; 78(2): 307-313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30178461

RESUMO

BACKGROUND: Neural tube defects are congenital malformations of the central nervous system. Genetic predisposition and some environmental factors play an important role in the development of neural tube defects. This study aimed to investigate the effects of diclofenac sodium on the neural tube development in a chick embryo model that corresponds to the first month of vertebral deve- lopment in mammals. MATERIALS AND METHODS: Seventy-five fertile, specific pathogen-free eggs were incubated for 28 h and were divided into five groups of 15 eggs each. Diclofenac sodium was administered via the sub-blastodermic route at this stage. Incubation was continued till the end of the 48th h. All eggs were then opened and embryos were dissected from embryonic membranes and evaluated morphologically and histopathologically. RESULTS: It was determined that the use of increasing doses of diclofenac sodium led to defects of midline closure in early chicken embryos. There were statistically significant differences in neural tube positions (open or close) among the groups. In addition; crown-rump length, somite number were significantly decreased in high dose experimental groups compared with control group. CONCLUSIONS: This study showed that development of neurons is affected in chi- cken embryos after administration of diclofenac sodium. The exact teratogenic mechanism of diclofenac sodium is not clear; therefore it should be investigated.


Assuntos
Diclofenaco/efeitos adversos , Tubo Neural/embriologia , Animais , Embrião de Galinha , Desenvolvimento Embrionário/efeitos dos fármacos , Tubo Neural/efeitos dos fármacos , Tubo Neural/patologia , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/patologia , Estatística como Assunto
4.
Congenit Anom (Kyoto) ; 57(5): 138-141, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28603928

RESUMO

Folate functions as a coenzyme to transfer one-carbon units that are necessary for deoxythymidylate synthesis, purine synthesis, and various methylation reactions. Ingested folate becomes a functional molecule through intestinal absorption, circulation, transport to cells, and various modifications to its structure. Associations between nutritional folate status and chronic diseases such as cardiovascular disease, cancer, and cognitive dysfunction have been reported. It has also been reported that maternal folate nutritional status is related to the risk of neural tube defects (NTDs) in the offspring. It has also been recommended that folate be consumed in the diet to promote the maintenance of good health. To reduce the risk of NTDs, supplementation with folic acid (a synthetic form of folate) during the periconceptional period has also been recommended. This paper describes the basic features and nutritional role of folate.


Assuntos
Suplementos Nutricionais , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/metabolismo , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/metabolismo , Embrião de Mamíferos , Feminino , Feto , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Humanos , Redes e Vias Metabólicas/fisiologia , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Defeitos do Tubo Neural/diagnóstico , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Recomendações Nutricionais
5.
Birth Defects Res ; 109(2): 81-91, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28008752

RESUMO

BACKGROUND: Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS: We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS: High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION: Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Proteínas de Transporte de Cátions/genética , Suplementos Nutricionais , Deficiência de Ácido Fólico/dietoterapia , Ácido Fólico/administração & dosagem , Ferro/administração & dosagem , Defeitos do Tubo Neural/prevenção & controle , Animais , Proteínas de Transporte de Cátions/deficiência , Cruzamentos Genéticos , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Deleção de Genes , Humanos , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Transgênicos , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Penetrância , Gravidez , Prosencéfalo/anormalidades , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo
6.
Birth Defects Res ; 109(2): 68-80, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-27324558

RESUMO

Susceptibility to neural tube defects (NTDs), such as anencephaly and spina bifida is influenced by genetic and environmental factors including maternal nutrition. Maternal periconceptional supplementation with folic acid significantly reduces the risk of an NTD-affected pregnancy, but does not prevent all NTDs, and "folic acid non-responsive" NTDs continue to occur. Similarly, among mouse models of NTDs, some are responsive to folic acid but others are not. Among nutritional factors, inositol deficiency causes cranial NTDs in mice while supplemental inositol prevents spinal and cranial NTDs in the curly tail (Grhl3 hypomorph) mouse, rodent models of hyperglycemia or induced diabetes, and in a folate-deficiency induced NTD model. NTDs also occur in mice lacking expression of certain inositol kinases. Inositol-containing phospholipids (phosphoinositides) and soluble inositol phosphates mediate a range of functions, including intracellular signaling, interaction with cytoskeletal proteins, and regulation of membrane identity in trafficking and cell division. Myo-inositol has been trialed in humans for a range of conditions and appears safe for use in human pregnancy. In pilot studies in Italy and the United Kingdom, women took inositol together with folic acid preconceptionally, after one or more previous NTD-affected pregnancies. In nonrandomized cohorts and a randomized double-blind study in the United Kingdom, no recurrent NTDs were observed among 52 pregnancies reported to date. Larger-scale fully powered trials are needed to determine whether supplementation with inositol and folic acid would more effectively prevent NTDs than folic acid alone. Birth Defects Research 109:68-80, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Suplementos Nutricionais , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/administração & dosagem , Inositol/administração & dosagem , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/efeitos dos fármacos , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Humanos , Fosfatos de Inositol/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Tubo Neural/anormalidades , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Fosfatidilinositóis/metabolismo , Gravidez
7.
Birth Defects Res ; 109(2): 106-119, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-27860192

RESUMO

BACKGROUND: Valproic acid (VPA) is prescribed therapeutically for multiple conditions, including epilepsy. When taken during pregnancy, VPA is teratogenic, increasing the risk of several birth and developmental defects including neural tube defects (NTDs). The mechanism by which VPA causes NTDs remains controversial and how VPA interacts with folic acid (FA), a vitamin commonly recommended for the prevention of NTDs, remains uncertain. We sought to address both questions by applying untargeted metabolite profiling analysis to neural tube closure (NTC) stage mouse embryos. METHODS: Pregnant SWV dams on either a 2 ppm or 10 ppm FA supplemented diet were injected with a single dose of VPA on gestational day E8.5. On day E9.5, the mouse embryos were collected and evaluated for NTC status. Liquid chromatography coupled to mass spectrometry metabolomics analysis was performed to compare metabolite profiles of NTD-affected VPA-exposed whole mouse embryos with profiles from embryos that underwent normal NTC from control dams. RESULTS: NTDs were observed in all embryos from VPA-treated dams and penetrance was not diminished by dietary FA supplementation. The most profound metabolic perturbations were found in the 10ppm FA VPA-exposed mouse embryos, compared with the other three treatment groups. Affected metabolites included amino acids, nucleobases and related phosphorylated nucleotides, lipids, and carnitines. CONCLUSION: Maternal VPA treatment markedly perturbed purine and pyrimidine metabolism in E9.5 embryos. In combination with a high FA diet, VPA treatment resulted in gross metabolic changes, likely caused by a multiplicity of mechanisms, including an apparent disruption of mitochondrial beta-oxidation. Birth Defects Research 109:106-119, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Suplementos Nutricionais , Ácido Fólico/administração & dosagem , Defeitos do Tubo Neural/metabolismo , Neurulação/efeitos dos fármacos , Teratogênicos/toxicidade , Ácido Valproico/toxicidade , Aminoácidos/metabolismo , Animais , Carnitina/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Lipídeos/análise , Masculino , Metaboloma , Camundongos , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/patologia , Penetrância , Gravidez , Purinas/metabolismo , Pirimidinas/metabolismo
8.
Int. j. morphol ; 34(2): 732-741, June 2016. ilus
Artigo em Espanhol | LILACS | ID: lil-787062

RESUMO

El ácido valproico (VPA) es el principal anticonvulsivante utilizado contra la epilepsia durante la gestación. Sin embargo, en etapas iniciales del embarazo actúa como teratógeno y ocasiona malformaciones como fisura labio-palatina, alteraciones en el desarrollo genital y espina bífida, siendo esta última la más frecuente. Esto se produce debido al aumento de especies reactivas de oxígeno, pudiendo contrarrestarse administrando vitamina E. El objetivo fue determinar si la vitamina E disminuye el daño en tubo neural y médula espinal de embriones y fetos de ratonas expuestas a VPA. Se conformaron 8 grupos de animales. A los 8 días post-fecundación se les administró a los grupos 1 y 5 suero fisiológico 0,3 mL; grupos 2 y 6 VPA 600 mg/Kg; grupos 3 y 7 VPA 600 mg/Kg y vitamina E 200 UI/Kg; grupos 4 y 8 vitamina E 200 UI/kg. A los 12 días post-fecundación, se sacrificaron los grupos 1, 2, 3 y 4, y a los 17 días los restantes grupos. Los embriones fueron procesados y teñidos con cresil violeta, observándose cortes histológicos a nivel cervical, torácico y lumbar. Los grupos tratados con vitamina E presentaron menor cantidad de neuroblastos y motoneuronas, pero de tamaño mayor en comparación al grupo tratado con VPA (p<0,05), siendo similares a los grupos controles. Al comparar el tubo neural y médula espinal en los distintos niveles (cervical, torácico y lumbar), no hubo diferencias estadísticamente significativas. La administración prenatal de vitamina E disminuye los defectos en tubo neural y médula espinal de embriones de 12 y 17 días de gestación sometidos a VPA.


Valproic Acid (VPA) is the main anticonvulsant used for epilepsy throughout the gestation period. However, when used at early stages of pregnancy, it acts as a tetarogenic agent, causing congenital malformations such as cleft-lip and/or cleft palate, abnormal genital development and spina bifida, being the latter the most frequent. This is the result of the increase of reactive oxygen species, which can be countered with the supplementation of vitamin E. The aim was determine if vitamin E minimizes the damage to the neural tube and spinal cord of mice embryos and fetuses previously exposed to VPA. Eight groups of mice were constituted. Eight days post fertilization, groups 1 and 5 were administered 0,3 ml of saline solution; groups 2 and 6 600mg/Kg of VPA, groups 3 and 7 600mg/Kg of VPA and 200UI/Kg of Vitamin E; groups 4 and 8 200 UI/Kg of Vitamin E. 12 days after fertilization, groups 1, 2, 3 and 4 were euthanized, whereas in the case of the remaining groups, the same process was performed 17 days after fertilization. The embryos were stained with cresyl violet, thus enabling the observation of histological sections at cervical, thoracic and lumbar levels. Groups supplied with vitamin E presented a lower amount of neuroblasts and motoneurons. However, these elements were bigger in size compared to the group treated with VPA (p<0,05), being these results similar to those obtained with the control groups. When comparing the neural tube and spinal cord at different levels (cervical, thoracic and lumbar), no statistically significant differences were found. It was determined that prenatal administration of vitamin E lessens the damage to the neural tube and spinal cord of mice embryos of 12 and 17 days of gestation previously exposed to VPA.


Assuntos
Animais , Feminino , Camundongos , Tubo Neural/efeitos dos fármacos , Tubo Neural/patologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Vitamina E/administração & dosagem , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/embriologia , Doenças da Medula Espinal/induzido quimicamente , Doenças da Medula Espinal/embriologia , Ácido Valproico/toxicidade
9.
Biochem Biophys Res Commun ; 467(2): 179-84, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-26453010

RESUMO

Maternal diabetes-induced birth defects remain a significant health problem. Studying the effect of natural compounds with antioxidant properties and minimal toxicities on diabetic embryopathy may lead to the development of new and safe dietary supplements. Punicalagin is a primary polyphenol found in pomegranate juice, which possesses antioxidant, anti-inflammatory and anti-tumorigenic properties, suggesting a protective effect of punicalagin on diabetic embryopathy. Here, we examined whether punicalagin could reduce high glucose-induced neural tube defects (NTDs), and if this rescue occurs through blockage of cellular stress and caspase activation. Embryonic day 8.5 (E8.5) mouse embryos were cultured for 24 or 36 h with normal (5 mM) glucose or high glucose (16.7 mM), in presence or absence of 10 or 20 µM punicalagin. 10 µM punicalagin slightly reduced NTD formation under high glucose conditions; however, 20 µM punicalagin significantly inhibited high glucose-induced NTD formation. Punicalagin suppressed high glucose-induced lipid peroxidation marker 4-hydroxynonenal, nitrotyrosine-modified proteins, and lipid peroxides. Moreover, punicalagin abrogated endoplasmic reticulum stress by inhibiting phosphorylated protein kinase ribonucleic acid (RNA)-like ER kinase (p-PERK), phosphorylated inositol-requiring protein-1α (p-IRE1α), phosphorylated eukaryotic initiation factor 2α (p-eIF2α), C/EBP-homologous protein (CHOP), binding immunoglobulin protein (BiP) and x-box binding protein 1 (XBP1) mRNA splicing. Additionally, punicalagin suppressed high glucose-induced caspase 3 and caspase 8 cleavage. Punicalagin reduces high glucose-induced NTD formation by blocking cellular stress and caspase activation. These observations suggest punicalagin supplements could mitigate the teratogenic effects of hyperglycemia in the developing embryo, and possibly prevent diabetes-induced NTDs.


Assuntos
Antioxidantes/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Taninos Hidrolisáveis/farmacologia , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Aldeídos/metabolismo , Animais , Caspases/genética , Caspases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Endorribonucleases/genética , Endorribonucleases/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Regulação da Expressão Gênica , Glucose , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/patologia , Fosforilação , Gravidez , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição de Fator Regulador X , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
10.
Am J Clin Nutr ; 101(4): 860-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25833982

RESUMO

BACKGROUND: Folic acid prevents neural tube closure defects (NTDs), but the causal metabolic pathways have not been established. Serine hydroxymethyltransferase 1 (SHMT1) is an essential scaffold protein in folate-dependent de novo thymidylate synthesis in the nucleus. SHMT1-deficient mice provide a model to investigate folic acid-responsive NTDs wherein disruption of de novo thymidylate synthesis impairs neural tube closure. OBJECTIVE: We examined the effects of maternal supplementation with the pyrimidine nucleosides uridine, thymidine, or deoxyuridine with and without folate deficiency on NTD incidence in the Shmt1 mouse model. DESIGN: Shmt1(+/+) and Shmt1(-/-) female mice fed folate-replete or folate-deficient diets and supplemented with uridine, thymidine, or deoxyuridine were bred, and litters (n = 10-23 per group) were examined for the presence of NTDs. Biomarkers of impaired folate status and metabolism were measured, including plasma nucleosides, hepatic uracil content, maternal plasma folate concentrations, and incorporation of nucleoside precursors into DNA. RESULTS: Shmt1(+/-) and Shmt1(-/-) embryos from dams fed the folate-deficient diet were susceptible to NTDs. No NTDs were observed in litters from dams fed the folate-deficient diet supplemented with deoxyuridine. Surprisingly, uridine supplementation increased NTD incidence, independent of embryo genotype and dietary folic acid. These dietary nucleosides did not affect maternal hepatic uracil accumulation in DNA but did affect plasma folate concentrations. CONCLUSIONS: Maternal deoxyuridine supplementation prevented NTDs in dams fed the folate-deficient diet, whereas maternal uridine supplementation increased NTD incidence, independent of folate and embryo genotype. These findings provide new insights into the metabolic impairments and mechanisms of folate-responsive NTDs resulting from decreased Shmt1 expression.


Assuntos
Desoxiuridina/administração & dosagem , Ácido Fólico/administração & dosagem , Defeitos do Tubo Neural/tratamento farmacológico , Uridina/administração & dosagem , Uridina/efeitos adversos , Animais , Desoxiuridina/sangue , Modelos Animais de Doenças , Feminino , Ácido Fólico/sangue , Deficiência de Ácido Fólico/tratamento farmacológico , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Células HeLa , Humanos , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Tubo Neural/efeitos dos fármacos , Defeitos do Tubo Neural/sangue , Defeitos do Tubo Neural/etiologia , Gravidez , Timidina/administração & dosagem , Timidina/efeitos adversos , Timidina/sangue , Uracila/metabolismo , Uridina/sangue
11.
Food Chem Toxicol ; 50(12): 4364-74, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22963837

RESUMO

Arsenic passes through the placenta and accumulates in the neuroepithelium of embryo, whereby inducing congenital malformations such as neural tube defects (NTDs) in animals. Choline (CHO), a methyl-rich nutrient, functions as a methyl donor to participate in methyl group metabolism. Arsenic methylation has been regarded as a detoxification process and choline (CHO) is the major source of methyl-groups. However, whether CHO intake reverses the abnormal embryo development induced by sodium arsenite (SA) and the relationship between CHO intake and arsenite-induced NTDs are still unclear. In this study, we used chick embryos as animal model to investigate the effects of SA and CHO supplementation on the early development of nervous system. Our results showed that the administration of SA led to reduction in embryo viability, embryo body weight and extraembryonic vascular area, accompanied by a significantly increased incidence of the failed closure of the caudal end of the neural tube. CHO, at low dose (25 µg/µL), reversed the decrease in embryo viability and the increase in the failed closure of the caudal end of the neural tube, which were induced by SA. In addition, CHO (25 µg/µL) inhibited not only the SA-induced cell apoptosis by up-regulating Bcl-2 level, but also the global DNA methylation by increasing the expressions of DNMT1 and DNMT3a. However, less significant difference was found between the embryos co-treated with SA and CHO (50 µg/µL) and the ones treated with SA alone. Taken together, these findings suggest that low dose CHO could protect chick embryos from arsenite-induced NTDs by a possible mechanism related to the methyl metabolism.


Assuntos
Arsenitos/toxicidade , Colina/farmacologia , Defeitos do Tubo Neural/patologia , Compostos de Sódio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Embrião de Galinha , Cricetinae , Metilação de DNA , Relação Dose-Resposta a Droga , Desenvolvimento Embrionário/efeitos dos fármacos , Modelos Animais , Tubo Neural/efeitos dos fármacos , Tubo Neural/patologia , Defeitos do Tubo Neural/induzido quimicamente , Regulação para Cima , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
12.
Hum Mol Genet ; 20(18): 3678-83, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21693562

RESUMO

Neural tube defects (NTDs), a common birth defect in humans, result from the failure of the embryonic neural tube (NT) to close properly. NT closure is a complex, poorly understood morphogenetic process influenced by genes and environment. The most effective environmental influence in decreasing the risk for NTDs is folic acid (FA) fortification and supplementation, and these findings led to the recommendation of periconceptual FA intake and mandatory fortification of the US grain supply in 1998. To explore the relationship between genetics and responsiveness to FA supplementation, we used five mouse NTDs models-Zic2, Shroom3, Frem2, Grhl2 (Grainyhead-like 2) and L3P (Line3P)-and a long-term generational FA supplementation scheme. Contrary to expectations, we find that three genetic mutants respond adversely to FA supplementation with increased incidence of NTDs in homozygous mutants, occurrence of NTDs in heterozygous embryos and embryonic lethality prior to NT closure. Because of these unexpected responses, we examined NTD risk after short-term FA supplementation. Our results indicate that, for the same genetic allele, NTD risk can depend on the length of FA exposure. Our data indicate that, depending on the gene mutation, FA supplementation may adversely influence embryonic development and NT closure.


Assuntos
Suplementos Nutricionais/efeitos adversos , Ácido Fólico/efeitos adversos , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/mortalidade , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Animais , Modelos Animais de Doenças , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/genética , Gravidez , Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA