Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Nutr ; 149(2): 295-303, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30689919

RESUMO

BACKGROUND: The risk of neural tube defect (NTD)-affected pregnancies is reduced with adequate folic acid intake during early pregnancy. However, NTDs have been observed among offspring of women with adequate folic acid intake. Some of these women are possibly not absorbing enough folic acid. Because lactase deficiency can lead to poor nutrient absorption, we hypothesized that lactase-deficient women will be at increased risk of having offspring with NTDs. OBJECTIVE: We examined the association between maternal rs4988235 (a lactase deficiency genetic marker) and NTDs in offspring. METHODS: We conducted a case-control study using data from the National Birth Defects Prevention Study, United States, 1997-2009, restricting to non-Hispanic white (NHW) and Hispanic women. Cases were women with an offspring with an NTD (n = 378 NHW, 207 Hispanic), and controls were women with an offspring without a birth defect (n = 461 NHW, 165 Hispanic). Analyses were conducted separately by race/ethnicity, using logistic regression. Women with the CC genotype were categorized as being lactase deficient. To assess potential effect modification, analyses were stratified by lactose intake, folic acid supplementation, dietary folate, and diet quality. RESULTS: Among NHW women, the odds of being lactase deficient were greater among cases compared with controls (OR: 1.37; 95% CI: 1.02, 1.82). Among Hispanic women, the odds of being lactase deficient were significantly lower among cases compared with controls (OR: 0.50, 95% CI: 0.33, 0.77). The association differed when stratified by lactose intake in NHW women (higher odds among women who consumed ≥12 g lactose/1000 kcal) and by dietary folate in Hispanic women (opposite direction of associations). The association did not differ when stratified by folic acid supplementation or diet quality. CONCLUSIONS: Our findings suggest that maternal lactase deficiency is associated with NTDs in offspring. However, we observed opposite directions of effect by race/ethnicity that could not be definitively explained.


Assuntos
Predisposição Genética para Doença , Lactase/genética , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único , Adulto , Estudos de Casos e Controles , Ácido Fólico/administração & dosagem , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/complicações , Marcadores Genéticos , Genótipo , Hispânico ou Latino , Humanos , Lactase/deficiência , Mães , Defeitos do Tubo Neural/enzimologia , Razão de Chances , Estados Unidos , Adulto Jovem
2.
Prenat Diagn ; 39(1): 3-9, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30474229

RESUMO

Published studies indicate the MTHFR C677T and A1298C polymorphisms are associated with abnormal homocysteine levels, which may cause various pregnancy complications and birth defects. However, the results obtained from different studies have been inconsistent. Therefore, this meta-analysis explores the association between MTHFR polymorphisms and birth defects and adverse pregnancy outcomes. The PubMed, ScienceDirect, Embase, and China Biology Medicine literature databases and ClinicalTrials were searched. Analyses of public bias, meta-regression, subgroups, and sensitivity were used to ensure the robustness of our results. MTHFR C677T was significantly associated with recurrent pregnancy loss in developing countries (odds ratio [OR], 1.34; 95% confidence interval [CI], 1.20-1.50) but not in developed countries (OR, 0.87; 95% CI, 0.68-1.11). No significant relationship was found between MTHFR A1298C and recurrent pregnancy loss (OR, 1.04; 95% CI, 0.93-1.18). MTHFR C677T and A1298C were not associated with preeclampsia (OR, 1.06; 95% CI, 0.97-1.16 and OR, 1.16; 95% CI, 0.97-1.39, respectively), and C677T was not associated with placental abruption (OR, 1.03; 95% CI, 0.87-1.21), intrauterine growth retardation (OR, 1.02; 95% CI, 0.90-1.15), or congenital heart disease (OR, 1.05; 95% CI, 0.89-1.25). MTHFR C677T, but not A1298C, was associated with neural tube defects (OR, 1.24; 95% CI, 1.08-1.42) and Down syndrome (OR, 1.65; 95% CI, 1.39-1.95). CONCLUSION: Although MTHFR C677T and A1298C are significantly associated with some types of congenital defects and adverse pregnancy outcomes, the impact of these polymorphisms is moderate.


Assuntos
Anormalidades Congênitas/enzimologia , Anormalidades Congênitas/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo Genético , Resultado da Gravidez/genética , Aborto Habitual/enzimologia , Aborto Habitual/genética , Ensaios Clínicos como Assunto , Países em Desenvolvimento , Síndrome de Down/enzimologia , Síndrome de Down/genética , Feminino , Retardo do Crescimento Fetal/enzimologia , Retardo do Crescimento Fetal/genética , Humanos , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/genética , Razão de Chances , Gravidez
3.
Medicina (Kaunas) ; 55(5)2019 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-31086097

RESUMO

Background and objectives: The pathophysiology of tethered cord syndrome (TCS) in children is not well elucidated. An inelastic filum terminale (FT) is the main factor underlying the stretching of the spinal cord in TCS. Our study aimed to investigate the expression of glutathione-S-transferase (GST) in children and fetal FT samples in order to understand the relationship between this enzyme expression and the development of TCS. Materials and Methods: FT samples were obtained from ten children with TCS (Group 1) and histological and immunohistochemical examinations were performed. For comparison, FT samples from fifteen normal human fetuses (Group 2) were also analyzed using the same techniques. Statistical comparison was made using a Chi-square test. Results: Positive GST-sigma expression was detected in eight (80%) of 10 samples in Group 1. The positive GST-sigma expression was less frequent in nine (60%) of 15 samples from Group 2. No statistically significant difference was detected between the two groups (p = 0.197). Conclusions: Decreased FT elasticity in TCS may be associated with increased GST expression in FT. More prospective studies are needed to clarify the mechanism of the GST-TCS relationship in children.


Assuntos
Glutationa/sangue , Defeitos do Tubo Neural/enzimologia , Cauda Equina , Distribuição de Qui-Quadrado , Pré-Escolar , Feminino , Glutationa/análise , Humanos , Lactente , Masculino , Defeitos do Tubo Neural/sangue , Estudos Prospectivos , Transferases/análise , Transferases/sangue
4.
Birth Defects Res A Clin Mol Teratol ; 100(1): 13-21, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24307569

RESUMO

BACKGROUND: Maternal exposure to polycyclic aromatic hydrocarbons (PAHs) has been associated with the risk of fetal neural tube defects (NTDs). Whether maternal genetic variants related to PAH metabolism contribute to the development of fetal NTDs remains unclear. METHODS: We conducted a case-control study in a Chinese population to examine the association of selected maternal genetic variants of phase II enzymes involved in the elimination of the metabolic intermediates of these chemicals with fetal NTD risk, and to evaluate possible interaction of the genetic variant and maternal exposure to indoor air pollution from coal combustion and smoking (IAPCC). Blood samples were collected from 534 NTD case mothers and 534 control mothers and assayed for 12 polymorphisms of 5 genes encoding phase II enzymes. RESULTS: We found that the rs9282861 GG genotype of SULT1A1 was associated with an elevated risk of total NTDs (odds ratio [OR] = 2.12, 95% confidence interval [CI]: 1.49-3.00), compared with the GA genotype. The SULT1A1 rs9282861 variant showed a significant additive interaction with maternal exposure to IAPCC for NTD risk, with a relative excess risk of interaction of 1.20 (95% CI 0.23-2.18), and the OR for the joint effect of high-level IAPCC exposure and the GG genotype was 8.37 (95% CI: 3.63-19.28). CONCLUSION: Maternal SULT1A1 polymorphism is associated with the risk of fetal NTDs, and has an additive-scale interaction with maternal IAPCC exposure for NTD risk.


Assuntos
Arilsulfotransferase/genética , Exposição Materna/efeitos adversos , Desintoxicação Metabólica Fase II/genética , Defeitos do Tubo Neural/genética , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Polimorfismo de Nucleotídeo Único , Adulto , Arilsulfotransferase/metabolismo , Estudos de Casos e Controles , China , Feminino , Feto , Interação Gene-Ambiente , Predisposição Genética para Doença , Humanos , Masculino , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/etiologia , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Fatores de Risco , População Rural , Smog/efeitos adversos
5.
Birth Defects Res A Clin Mol Teratol ; 100(1): 22-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24382678

RESUMO

UNLABELLED: Maternal tea consumption was reported to increase the risk of fetal neural tube defects (NTDs). Catechol-O-methyltransferase (COMT) may be involved in the metabolism of polyphenolic methylation of tea, thus influence the risk of fetal NTDs. METHODS: A total of 576 fetuses or newborns with NTDs and 594 healthy newborns were included in the case-control study. Information on maternal tea consumption, sociodemographic characteristics, reproductive history, and related behavior was collected through face-to-face interviews. Maternal blood samples were collected to examine polymorphisms in COMT, and the possible interaction of COMT and tea consumption was analyzed. RESULTS: After controlling for potential confounders, homozygotes of rs737865 showed an elevated risk for total NTDs (odds ratio [OR] = 2.04, 95% confidence interval [CI], 1.24-3.35) and for the anencephaly subtype (OR = 1.99, 95% CI, 1.17-3.39). The CC genotype of rs4633 was positively associated with the overall risk of NTDs (OR = 3.66, 95% CI, 1.05-12.83). Heterozygotes for rs4680 were associated with a decreased risk of spina bifida (OR = 0.71, 95% CI, 0.51-0.98). The COMT rs4680 A allele was negatively related with the risk of spina bifida, with adjusted OR = 0.64 (95% CI, 0.45-0.89). An interaction between tea consumption (1 to 2 cups/day) and the rs4680AA/AG genotype was found in the spina bifida subtype (Pinteraction = .08). CONCLUSION: Several COMT variants were associated with elevated risk of NTDs in a Chinese population. Maternal tea consumption may be associated with an increased risk for fetal NTDs in genetically susceptible subgroups.


Assuntos
Anencefalia/genética , Catecol O-Metiltransferase/genética , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único , Disrafismo Espinal/genética , Chá/efeitos adversos , Adulto , Anencefalia/induzido quimicamente , Anencefalia/enzimologia , Estudos de Casos e Controles , Catecol O-Metiltransferase/metabolismo , China , Feminino , Feto , Predisposição Genética para Doença , Humanos , Masculino , Exposição Materna/efeitos adversos , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/enzimologia , Razão de Chances , Polifenóis/toxicidade , Fatores de Risco , População Rural , Disrafismo Espinal/induzido quimicamente , Disrafismo Espinal/enzimologia
6.
Proc Natl Acad Sci U S A ; 106(24): 9831-5, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19482943

RESUMO

Inositol 1,3,4-trisphosphate 5/6-kinase (ITPK1) is a key regulatory enzyme at the branch point for the synthesis of inositol hexakisphosphate (IP(6)), an intracellular signaling molecule implicated in the regulation of ion channels, endocytosis, exocytosis, transcription, DNA repair, and RNA export from the nucleus. IP(6) also has been shown to be an integral structural component of several proteins. We have generated a mouse strain harboring a beta-galactosidase (betagal) gene trap cassette in the second intron of the Itpk1 gene. Animals homozygous for this gene trap are viable, fertile, and produce less ITPK1 protein than wild-type and heterozygous animals. Thus, the gene trap represents a hypomorphic rather than a null allele. Using a combination of immunohistochemistry, in situ hybridization, and betagal staining of mice heterozygous for the hypomorphic allele, we found high expression of Itpk1 in the developing central and peripheral nervous systems and in the paraxial mesoderm. Examination of embryos resulting from homozygous matings uncovered neural tube defects (NTDs) in some animals and axial skeletal defects or growth retardation in others. On a C57BL/6 x 129(P2)Ola background, 12% of mid-gestation embryos had spina bifida and/or exencephaly, whereas wild-type animals of the same genetic background had no NTDs. We conclude that ITPK1 is required for proper development of the neural tube and axial mesoderm.


Assuntos
Defeitos do Tubo Neural/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Desenvolvimento Embrionário , Masculino , Camundongos , Camundongos Transgênicos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Birth Defects Res A Clin Mol Teratol ; 91(9): 848-56, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21770021

RESUMO

BACKGROUND: Neural tube defects (NTDs) are among the most common severe congenital malformations, representing a long-term public health burden in India. A deranged one-carbon metabolism and genes regulating this metabolism have been linked to NTDs. Vitamin B(12) deficiency is reported to be more prevalent than folate deficiency in the Indian population. We investigated the role of maternal nutritional and genetic markers related to one-carbon metabolism in the etiology of NTDs. METHODS: We conducted a multicenter case-control study to compare plasma folate, vitamin B(12) , homocysteine and holo-transcobalamin levels, and polymorphisms in methylenetetrahydrofolate reductase (MTHFR, 677C>T, 1298A>C, 1781G>A and 236+724A>G) and transcobalamin (TCN2, 776C>G) genes, in 318 women with NTD-affected offspring (cases) and 702 women with apparently healthy offspring (controls). The samples were collected at diagnosis in cases and at delivery in controls. RESULTS: We observed a significant association of high maternal plasma homocysteine concentrations with NTDs in the offspring (p = 0.026). There was no association of maternal folate or B(12) levels with NTDs (p > 0.05) but low maternal holo-transcobalamin predicted strong risk of NTDs in the offspring (p = 0.003). The commonly associated maternal polymorphism 677C>T in the MTHFR gene did not predict risk of NTDs in the offspring (p > 0.05) and 1298A>C and 1781G>A polymorphisms in MTHFR were protective (p = 0.024 and 0.0004 respectively). Maternal 776C>G polymorphism in TCN2 was strongly predictive of NTD in the offspring (p = 0.006). CONCLUSION: Our study has demonstrated a possible role for maternal B(12) deficiency in the etiology of NTDs in India over and above the well-established role of folate deficiency.


Assuntos
Homocisteína/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2) , Defeitos do Tubo Neural , Polimorfismo de Nucleotídeo Único , Transcobalaminas , Adulto , Estudos de Casos e Controles , Feminino , Ácido Fólico/genética , Ácido Fólico/metabolismo , Predisposição Genética para Doença , Humanos , Índia/epidemiologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/genética , Transcobalaminas/genética , Transcobalaminas/metabolismo , Vitamina B 12/genética , Vitamina B 12/metabolismo , Deficiência de Vitamina B 12/enzimologia , Deficiência de Vitamina B 12/epidemiologia , Deficiência de Vitamina B 12/genética
8.
Genet Mol Res ; 10(4): 2424-9, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-22002135

RESUMO

Cystathionine beta synthase gene (CßS) catalyzes the condensation of homocysteine with serine, forming cystathionine by the transsulfuration pathway. Disruption of CßS enzyme activity due to defective folic acid metabolism increases the risk factor for neural tube defects. We evaluated the CßS gene mutation in 25 children with neural tube defects (NTDs), including lumbosacral and thoracic myelomeningocele and open NTDs and mothers of cases, along with 25 healthy children and their mothers, serving as controls. Genomic DNA was isolated to assess the polymorphism of 852Ins68 in the CßS gene using PCR-RFLP analysis and nucleotide sequencing techniques. The 68-bp insertion was observed in one of the 25 NTD cases (lumbosacral myelomeningocele), and in two of the mothers of NTD cases. Statistical analysis was carried out using the Fischer exact probability test, which showed a lack of significance (P > 0.05), but the odds ratio of 2.08 with 95% confidence interval of 0.17-24.6 in NTDs mother was quite high because of the small sample size. However, the study was further extended to find out the involvement of specific nucleotide sequences, which again confirmed the 852Ins68 insertion and replacement of nucleotides (TCCAT to GGGG) in lumbosacral myelomeningocele (due to other category of NTDs), suggesting that it could be an independent risk factor for birth defects, including NTDs.


Assuntos
Cistationina beta-Sintase/genética , Meningomielocele/genética , Mutagênese Insercional , Defeitos do Tubo Neural/genética , Polimorfismo de Fragmento de Restrição , Adulto , Pré-Escolar , Cistationina beta-Sintase/metabolismo , Feminino , Humanos , Índia/epidemiologia , Lactente , Recém-Nascido , Masculino , Meningomielocele/enzimologia , Meningomielocele/epidemiologia , Defeitos do Tubo Neural/enzimologia , Reação em Cadeia da Polimerase/métodos , Prevalência , Fatores de Risco
9.
Birth Defects Res A Clin Mol Teratol ; 88(10): 883-94, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20842651

RESUMO

Retinoic acid (RA) is a pleiotropic derivative of vitamin A, or retinol, which is responsible for all of the bioactivity associated with this vitamin. The teratogenic influences of vitamin A deficiency and excess RA in rodents were first observed more than 50 years ago. Efforts over the last 15-20 years have refined these observations by defining the molecular mechanisms that control RA availability and signaling during murine embryonic development. This review will discuss our current understanding of the role of RA in teratogenesis, with specific emphasis on the essential function of the RA catabolic CYP26 enzymes in preventing teratogenic consequences caused by uncontrolled distribution of RA. Particular focus will be paid to the RA-sensitive tissues of the caudal and cranial regions, the limb, and the testis, and how genetic mutation of factors controlling RA distribution have revealed important roles for RA during embryogenesis.


Assuntos
Anormalidades Congênitas/enzimologia , Sistema Enzimático do Citocromo P-450/metabolismo , Desenvolvimento Embrionário , Tretinoína/metabolismo , Deficiência de Vitamina A/enzimologia , Animais , Anormalidades Congênitas/embriologia , Anormalidades Congênitas/metabolismo , Extremidades/embriologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/enzimologia , Gravidez , Ácido Retinoico 4 Hidroxilase , Teratogênicos/metabolismo , Testículo/embriologia , Deficiência de Vitamina A/embriologia , Deficiência de Vitamina A/metabolismo
10.
Mol Cell Biol ; 27(9): 3405-16, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17325035

RESUMO

Gcn5 was the first transcription-related histone acetyltransferase (HAT) to be identified. However, the functions of this enzyme in mammalian cells remain poorly defined. Deletion of Gcn5 in mice leads to early embryonic lethality with increased apoptosis in mesodermal lineages. Here we show that deletion of p53 allows Gcn5(-/-) embryos to survive longer, but Gcn5(-/-) p53(-/-) embryos still die in midgestation. Interestingly, embryos homozygous for point mutations in the Gcn5 catalytic domain survive significantly longer than Gcn5(-/-) or Gcn5(-/-) p53(-/-) mice. In contrast to Gcn5(-/-) embryos, Gcn5(hat/hat) embryos do not exhibit increased apoptosis but do exhibit severe cranial neural tube closure defects and exencephaly. Together, our results indicate that Gcn5 has important, HAT-independent functions in early development and that Gcn5 acetyltransferase activity is required for cranial neural tube closure in the mouse.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/enzimologia , Histona Acetiltransferases/deficiência , Histona Acetiltransferases/metabolismo , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Acetilação , Animais , Apoptose , Biomarcadores , Proteínas de Ciclo Celular/genética , Perda do Embrião , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Histona Acetiltransferases/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Defeitos do Tubo Neural/genética , Neurônios/metabolismo , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fatores de Transcrição de p300-CBP
11.
Fetal Diagn Ther ; 28(1): 34-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20516661

RESUMO

OBJECTIVE: To evaluate oxidative stress markers and prolidase activity in the amniotic fluid of fetuses with a neural tube defect (NTD) compared to the amniotic fluid of normal fetuses. METHODS: Amniotic fluid samples from 36 pregnancies of fetuses with NTD were compared with the amniotic fluid from 36 pregnancies of genetically normal fetuses for analysis of oxidative status and prolidase activity. The control group consisted of fetuses from pregnancies that were diagnosed as high-risk according to a 1st or 2nd trimester aneuploidy screening, but which were later confirmed to have normal results. An automated method was used to measure oxidative status. Prolidase enzyme activity was determined using a photometric method. RESULTS: Prolidase activity, total oxidant status and oxidative stress index of amniotic fluid from fetuses with NTD were significantly higher compared to controls (p < 0.001, p = 0.003 and p = 0.002, respectively), whereas total antioxidant capacity was significantly lower (p < 0.001). CONCLUSION: Levels of prolidase activity and oxidative stress are increased in the amniotic fluid of fetuses with NTD. These indicators may serve as diagnostic markers in this disease.


Assuntos
Líquido Amniótico/enzimologia , Dipeptidases/metabolismo , Feto/enzimologia , Defeitos do Tubo Neural/enzimologia , Feminino , Humanos , Oxirredução , Gravidez
12.
CNS Neurosci Ther ; 26(9): 902-912, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32329577

RESUMO

AIMS: Failure of neural tube closure resulting from excessive apoptosis leads to neural tube defects (NTDs). NADPH oxidase 4 (NOX4) is a critical mediator of cell growth and death, yet its role in NTDs has never been characterized. NOX4 is a potential target of miR-322, and we have previously demonstrated that miR-322 was involved in high glucose-induced NTDs. In this study, we investigated the effect of NOX4 on the embryonic neuroepithelium in NTDs and reveal a new regulatory mechanism for miR-322 that disrupts neurulation by ameliorating cell apoptosis. METHODS: All-trans-retinoic acid (ATRA)-induced mouse model was utilized to study NTDs. RNA pull-down and dual-luciferase reporter assays were used to confirm the interaction between NOX4 and miR-322. In mouse neural stem cells and whole-embryo culture, Western blot and TUNEL were carried out to investigate the effects of miR-322 and NOX4 on neuroepithelium apoptosis in NTD formation. RESULTS: NOX4, as a novel target of miR-322, was upregulated in ATRA-induced mouse model of NTDs. In mouse neural stem cells, the expression of NOX4 was inhibited by miR-322; still further, NOX4-triggered apoptosis was also suppressed by miR-322. Moreover, in whole-embryo culture, injection of the miR-322 mimic into the amniotic cavity attenuated cell apoptosis in NTD formation by silencing NOX4. CONCLUSION: miR-322/NOX4 plays a crucial role in apoptosis-induced NTD formation, which may provide a new understanding of the mechanism of embryonic NTDs and a basis for potential therapeutic target against NTDs.


Assuntos
Apoptose/fisiologia , Inativação Gênica/fisiologia , MicroRNAs/administração & dosagem , NADPH Oxidase 4/antagonistas & inibidores , NADPH Oxidase 4/biossíntese , Defeitos do Tubo Neural/enzimologia , Animais , Células Cultivadas , Desenvolvimento Embrionário/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , NADPH Oxidase 4/genética , Defeitos do Tubo Neural/diagnóstico por imagem , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/terapia , Resultado do Tratamento
13.
Hum Mutat ; 30(12): 1650-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19777576

RESUMO

Polymorphisms in folate-related genes have emerged as important risk factors in a range of diseases including neural tube defects (NTDs), cancer, and coronary artery disease (CAD). Having previously identified a polymorphism within the cytoplasmic folate enzyme, MTHFD1, as a maternal risk factor for NTDs, we considered the more recently identified mitochondrial paralogue, MTHFD1L, as a candidate gene for NTD association. We identified a common deletion/insertion polymorphism, rs3832406, c.781-6823ATT(7-9), which influences splicing efficiency and is strongly associated with NTD risk. Three alleles of rs3832406 were detected in the Irish population with varying numbers of ATT repeats: Allele 1 consists of ATT(7), whereas Alleles 2 and 3 consist of ATT(8) and ATT(9), respectively. Allele 2 of this triallelic polymorphism showed a decreased case risk as demonstrated by case-control logistic regression (P=0.002) and by transmission disequilibrium test (TDT) (P=0.001), whereas Allele 1 showed an increased case risk. Allele 3 showed no influence on NTD risk and represents the lowest frequency allele (0.15). Additional single nucleotide polymorphism (SNP) genotyping in the same genomic region provides additional supportive evidence of an association. We demonstrate that two of the three alleles of rs3832406 are functionally different and influence the splicing efficiency of the alternate MTHFD1L mRNA transcripts.


Assuntos
Processamento Alternativo/genética , Predisposição Genética para Doença , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único/genética , Sequência de Bases , Estudos de Casos e Controles , Frequência do Gene/genética , Haplótipos/genética , Humanos , Desequilíbrio de Ligação/genética , Antígenos de Histocompatibilidade Menor , Modelos Biológicos , Dados de Sequência Molecular
14.
Hum Genet ; 125(3): 247-56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19130090

RESUMO

Genetic variants in MTHFD1 (5,10-methylenetetrahydrofolate dehydrogenase/5,10-methenyltetrahydrofolate cyclohydrolase/ 10-formyltetrahydrofolate synthetase), an important folate metabolic enzyme, are associated with a number of common diseases, including neural tube defects (NTDs). This study investigates the promoter of the human MTHFD1 gene in a bid to understand how this gene is controlled and regulated. Following a combination of in silico and molecular approaches, we report that MTHFD1 expression is controlled by a TATA-less, Initiator-less promoter and transcription is initiated at multiple start sites over a 126 bp region. We confirmed the presence of three database polymorphisms (dbSNP) by direct sequencing of the upstream region (rs1076991 C > T, rs8010584 G > A, rs4243628 G > T), with a fourth (dbSNP rs746488 A > T) not found to be polymorphic in our population and no novel polymorphisms identified. We demonstrate that a common SNP rs1076991 C > T within the window of transcriptional initiation exerts a significant effect on promoter activity in vitro. We investigated this SNP as a potential risk factor for NTDs in a large homogenous Irish population and determined that it is not an independent risk factor, but, it does increase both case (chi (2) = 11.06, P = 0.001) and maternal (chi (2) = 6.68, P = 0.01) risk when allele frequencies were analysed in combination with the previously identified disease-associated p.R653Q (c.1958 G > A; dbSNP rs2236225) polymorphism. These results provide the first insight into how MTHFD1 is regulated and further emphasise its importance during embryonic development.


Assuntos
Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/genética , Regiões Promotoras Genéticas , Alelos , Sequência de Bases , Estudos de Casos e Controles , Biologia Computacional , Primers do DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Humanos , Recém-Nascido , Irlanda , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Gravidez , Fatores de Risco , Sítio de Iniciação de Transcrição
15.
Birth Defects Res A Clin Mol Teratol ; 85(6): 531-41, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19215022

RESUMO

BACKGROUND: Despite extensive research on mild methylenetetrahydrofolate reductase (MTHFR) deficiency and low dietary folate in different disorders, the association of these metabolic disturbances with a variety of congenital defects and pregnancy complications remains controversial. In this study we investigated the effects of MTHFR and dietary folate deficiency at 10.5 days post coitum (dpc) in our mouse model of mild MTHFR deficiency. METHODS: Mthfr +/+ and +/- female mice were fed a control or folic acid-deficient diet for 6 weeks, then mated with Mthfr +/- males. At 10.5 dpc, embryos were examined and placentae were collected for histologic evaluation. RESULTS: Maternal MTHFR and folate deficiencies resulted in increased developmental delays and smaller embryos. We also observed a low frequency of a variety of embryonic defects in the experimental groups, such as neural tube, heart looping, and turning defects; these results mimic the low incidence and multifactorial nature of these anomalies in humans. Folate-deficient mice also had increased embryonic losses and severe placental defects, including placental abruption and disturbed patterning of placental layers. Folate-deficient placentae had decreased ApoA-I expression, and there was a trend toward a negative correlation between ApoA-I expression with maternal homocysteine concentrations. CONCLUSIONS: Our study provides biological evidence linking maternal MTHFR and dietary folate deficiencies to adverse pregnancy outcomes in mice. It underscores the importance of folate not only in reducing the incidence of early embryonic defects, but also in the prevention of developmental delays and placental abnormalities that may increase susceptibility to other defects and to reproductive complications.


Assuntos
Suplementos Nutricionais , Desenvolvimento Embrionário , Ácido Fólico/farmacologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/deficiência , Placenta/anormalidades , Placenta/enzimologia , Complexo Vitamínico B/farmacologia , Animais , Apolipoproteína A-I/biossíntese , Perda do Embrião/enzimologia , Embrião de Mamíferos/enzimologia , Feminino , Cardiopatias Congênitas/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Defeitos do Tubo Neural/enzimologia , Gravidez
16.
Artigo em Inglês | MEDLINE | ID: mdl-19194987

RESUMO

Maternal diabetes causes neural tube defects in embryos, which are associated with increased apoptosis in the neuroepithelium. Many factors, including effector caspases, have been shown to be involved in the events. However, the key regulators have not been identified and the underlying mechanisms remain to be addressed. Caspase-8, an initiator caspase, has been shown to be altered in diabetic embryopathy, suggesting a role as an upstream apoptotic regulator. Using mouse embryos as a model system, this study demonstrates that caspase-8 is required for the production of hyperglycemia-associated embryonic malformations. Caspase-8 was shown to be expressed in the developing neural tube. Its activity, as evidenced by enhanced cleavage, was increased by hyperglycemia. These changes were associated with increased formation of the active cleavage of Bid. Inhibition of caspase-8 activity in high glucose-challenged embryos reduced the rate of embryonic malformation and this was associated with decreased apoptosis in the neuroepithelium of the neural tube. Inhibition of caspase-8 activity also reduced hyperglycemia-induced Bid activation and caspase-9 cleavage. These data suggest that caspase-8 may control diabetic embryopathy-associated apoptosis via regulation of the Bid-stimulated mitochondrion/caspase-9 pathway.


Assuntos
Anormalidades Induzidas por Medicamentos/enzimologia , Caspase 8/metabolismo , Diabetes Mellitus Experimental/enzimologia , Defeitos do Tubo Neural/enzimologia , Tubo Neural/enzimologia , Gravidez em Diabéticas/enzimologia , Anormalidades Induzidas por Medicamentos/etiologia , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Glucose/farmacologia , Processamento de Imagem Assistida por Computador , Camundongos , Tubo Neural/anormalidades , Defeitos do Tubo Neural/etiologia , Gravidez , Gravidez em Diabéticas/etiologia
17.
Am J Clin Nutr ; 109(3): 674-683, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30848279

RESUMO

BACKGROUND: The risk of neural tube defects (NTDs) is influenced by nutritional factors and genetic determinants of one-carbon metabolism. A key pathway of this metabolism is the vitamin B-12- and folate-dependent remethylation of homocysteine, which depends on methionine synthase (MS, encoded by MTR), methionine synthase reductase, and methylenetetrahydrofolate reductase. Methionine, the product of this pathway, is the direct precursor of S-adenosylmethionine (SAM), the universal methyl donor needed for epigenetic mechanisms. OBJECTIVES: This study aimed to evaluate whether the availability of vitamin B-12 and folate and the expression or activity of the target enzymes of the remethylation pathway are involved in NTD risk. METHODS: We studied folate and vitamin B-12 concentrations and activity, expression, and gene variants of the 3 enzymes in liver from 14 NTD and 16 non-NTD fetuses. We replicated the main findings in cord blood from pregnancies of 41 NTD fetuses compared with 21 fetuses with polymalformations (metabolic and genetic findings) and 375 control pregnancies (genetic findings). RESULTS: The tissue concentration of vitamin B-12 (P = 0.003), but not folate, and the activity (P = 0.001), transcriptional level (P = 0.016), and protein expression (P = 0.003) of MS were decreased and the truncated inactive isoforms of MS were increased in NTD livers. SAM was significantly correlated with MS activity and vitamin B-12. A gene variant in exon 1 of GIF (Gastric Intrinsic Factor gene) was associated with a dramatic decrease of liver vitamin B-12 in 2 cases. We confirmed the decreased vitamin B-12 in cord blood from NTD pregnancies. A gene variant of GIF exon 3 was associated with NTD risk. CONCLUSIONS: The decreased vitamin B-12 in liver and cord blood and decreased expression and activity of MS in liver point out the impaired remethylation pathway as hallmarks associated with NTD risk. We suggest evaluating vitamin B-12 in the nutritional recommendations for prevention of NTD risk beside folate fortification or supplementation.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Doenças Fetais/enzimologia , Fígado/metabolismo , Defeitos do Tubo Neural/enzimologia , Vitamina B 12/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Estudos de Casos e Controles , Feminino , Ferredoxina-NADP Redutase/genética , Ferredoxina-NADP Redutase/metabolismo , Doenças Fetais/genética , Doenças Fetais/metabolismo , Ácido Fólico/análise , Ácido Fólico/metabolismo , Idade Gestacional , Humanos , Fígado/química , Fígado/embriologia , Fígado/enzimologia , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Gravidez , Vitamina B 12/análise
18.
Biochim Biophys Acta ; 1773(8): 1341-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17306896

RESUMO

c-Jun N-terminal kinases (JNKs), also referred to as stress-activated kinases (SAPKs), were initially characterized by their activation in response to cell stress such as UV irradiation. JNK/SAPKs have since been characterized to be involved in proliferation, apoptosis, motility, metabolism and DNA repair. Dysregulated JNK signaling is now believed to contribute to many diseases involving neurodegeneration, chronic inflammation, birth defects, cancer and ischemia/reperfusion injury. In this review, we present our current understanding of JNK regulation and their involvement in homeostasis and dysregulation in human disease.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sequência de Aminoácidos , Animais , Doença/etiologia , Feminino , Perda Auditiva/enzimologia , Humanos , Inflamação/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/química , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fígado/irrigação sanguínea , Fígado/lesões , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Dados de Sequência Molecular , Neoplasias/enzimologia , Defeitos do Tubo Neural/enzimologia , Doenças Neurodegenerativas/enzimologia , Gravidez , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais
19.
Mol Cell Biol ; 25(24): 11113-21, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16314531

RESUMO

Sphingosine-1-phosphate (S1P), an important sphingolipid metabolite, regulates diverse cellular processes, including cell survival, growth, and differentiation. Here we show that S1P signaling is critical for neural and vascular development. Sphingosine kinase-null mice exhibited a deficiency of S1P which severely disturbed neurogenesis, including neural tube closure, and angiogenesis and caused embryonic lethality. A dramatic increase in apoptosis and a decrease in mitosis were seen in the developing nervous system. S1P(1) receptor-null mice also showed severe defects in neurogenesis, indicating that the mechanism by which S1P promotes neurogenesis is, in part, signaling from the S1P(1) receptor. Thus, S1P joins a growing list of signaling molecules, such as vascular endothelial growth factor, which regulate the functionally intertwined pathways of angiogenesis and neurogenesis. Our findings also suggest that exploitation of this potent neuronal survival pathway could lead to the development of novel therapeutic approaches for neurological diseases.


Assuntos
Vasos Sanguíneos/embriologia , Lisofosfolipídeos/metabolismo , Neovascularização Fisiológica , Sistema Nervoso/embriologia , Defeitos do Tubo Neural/genética , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Esfingosina/análogos & derivados , Animais , Apoptose/genética , Vasos Sanguíneos/anormalidades , Vasos Sanguíneos/enzimologia , Camundongos , Camundongos Knockout , Mitose/genética , Neovascularização Fisiológica/genética , Sistema Nervoso/enzimologia , Defeitos do Tubo Neural/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/metabolismo
20.
Mol Cell Biol ; 25(20): 8948-59, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16199873

RESUMO

Skeletal disorders and neural tube closure defects represent clinically significant human malformations. The signaling networks regulating normal skeletal patterning and neurulation are largely unknown. Targeted mutation of the active site lysine of MEK kinase 4 (MEKK4) produces a kinase-inactive MEKK4 protein (MEKK4(K1361R)). Embryos homozygous for this mutation die at birth as a result of skeletal malformations and neural tube defects. Hindbrains of exencephalic MEKK4(K1361R) embryos show a striking increase in neuroepithelial cell apoptosis and a dramatic loss of phosphorylation of MKK3 and -6, mitogen-activated protein kinase kinases (MKKs) regulated by MEKK4 in the p38 pathway. Phosphorylation of MAPK-activated protein kinase 2, a p38 substrate, is also inhibited, demonstrating a loss of p38 activity in MEKK4(K1361R) embryos. In contrast, the MEK1/2-extracellular signal-regulated kinase 1 (ERK1)/ERK2 and MKK4-Jun N-terminal protein kinase pathways were unaffected. The p38 pathway has been shown to regulate the phosphorylation and expression of the small heat shock protein HSP27. Compared to the wild type, MEKK4(K1361R) fibroblasts showed significantly reduced phosphorylation of p38 and HSP27, with a corresponding heat shock-induced instability of the actin cytoskeleton. Together, these data demonstrate MEKK4 regulation of p38 and that substrates downstream of p38 control cellular homeostasis. The findings are the first demonstration that MEKK4-regulated p38 activity is critical for neurulation.


Assuntos
Desenvolvimento Ósseo/fisiologia , MAP Quinase Quinase Quinase 4/deficiência , Defeitos do Tubo Neural/enzimologia , Animais , Apoptose , Sequência de Bases , Padronização Corporal/genética , Padronização Corporal/fisiologia , Desenvolvimento Ósseo/genética , DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Marcação de Genes , Humanos , MAP Quinase Quinase Quinase 4/genética , MAP Quinase Quinase Quinase 4/fisiologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/patologia , Fenótipo , Fosforilação , Gravidez , Rombencéfalo/anormalidades , Rombencéfalo/enzimologia , Rombencéfalo/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA