Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
CNS Neurosci Ther ; 26(9): 902-912, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32329577

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Silenciador del Gen/fisiología , MicroARNs/administración & dosificación , NADPH Oxidasa 4/antagonistas & inhibidores , NADPH Oxidasa 4/biosíntesis , Defectos del Tubo Neural/enzimología , Animales , Células Cultivadas , Desarrollo Embrionario/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , NADPH Oxidasa 4/genética , Defectos del Tubo Neural/diagnóstico por imagen , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/terapia , Resultado del Tratamiento
2.
Medicina (Kaunas) ; 55(5)2019 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-31086097

RESUMEN

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.


Asunto(s)
Glutatión/sangre , Defectos del Tubo Neural/enzimología , Cauda Equina , Distribución de Chi-Cuadrado , Preescolar , Femenino , Glutatión/análisis , Humanos , Lactante , Masculino , Defectos del Tubo Neural/sangre , Estudios Prospectivos , Transferasas/análisis , Transferasas/sangre
3.
Am J Clin Nutr ; 109(3): 674-683, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30848279

RESUMEN

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.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Enfermedades Fetales/enzimología , Hígado/metabolismo , Defectos del Tubo Neural/enzimología , Vitamina B 12/metabolismo , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Estudios de Casos y Controles , Femenino , Ferredoxina-NADP Reductasa/genética , Ferredoxina-NADP Reductasa/metabolismo , Enfermedades Fetales/genética , Enfermedades Fetales/metabolismo , Ácido Fólico/análisis , Ácido Fólico/metabolismo , Edad Gestacional , Humanos , Hígado/química , Hígado/embriología , Hígado/enzimología , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Embarazo , Vitamina B 12/análisis
4.
J Nutr ; 149(2): 295-303, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30689919

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad , Lactasa/genética , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple , Adulto , Estudios de Casos y Controles , Ácido Fólico/administración & dosificación , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Marcadores Genéticos , Genotipo , Hispánicos o Latinos , Humanos , Lactasa/deficiencia , Madres , Defectos del Tubo Neural/enzimología , Oportunidad Relativa , Estados Unidos , Adulto Joven
5.
Prenat Diagn ; 39(1): 3-9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30474229

RESUMEN

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.


Asunto(s)
Anomalías Congénitas/enzimología , Anomalías Congénitas/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Resultado del Embarazo/genética , Aborto Habitual/enzimología , Aborto Habitual/genética , Ensayos Clínicos como Asunto , Países en Desarrollo , Síndrome de Down/enzimología , Síndrome de Down/genética , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/genética , Humanos , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/genética , Oportunidad Relativa , Embarazo
7.
Reprod Toxicol ; 76: 17-25, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29258758

RESUMEN

Thymidylate synthase (TYMS) is a key enzyme in the de novo synthesis of 2'-deoxythymidine-5'-monophosphate (dTMP) from 2'-deoxyuridine-5'-monophosphate (dUMP). Our aim was to investigate the role of dTMP dysmetabolism via inhibition of TYMS by an inhibitor, 5-fluorouracil (5-FU) in the occurrence of neural tube defects (NTDs). We found that a high incidence of NTDs occurred after treatment with 5-FU at 12.5 mg/kg body weight. TYMS activity was significantly inhibited with decreased dTMP and accumulation of dUMP after 5-FU injection. The proliferation of neuroepithelial cells were markedly inhibited in NTDs compared with control. Expressions of proliferating cell nuclear antigen and phosphohistone H3 were significantly decreased in NTDs, while phosphorylated replication protein A2, P53 and Caspase3 were significantly increased in NTDs compared with control. These results indicated that inhibition of TYMS affected the balance between proliferation and apoptosis in neuroepithelial cells, which might shed some lights on the mechanisms involved in NTDs.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Defectos del Tubo Neural/enzimología , Tubo Neural/efectos de los fármacos , Timidilato Sintasa/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Nucleótidos de Desoxiuracil/metabolismo , Fluorouracilo/toxicidad , Ratones Endogámicos C57BL , Tubo Neural/embriología , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Células Neuroepiteliales/efectos de los fármacos , Células Neuroepiteliales/enzimología , Células Neuroepiteliales/patología , Timidina/análogos & derivados , Timidina/metabolismo
8.
Birth Defects Res ; 109(13): 1020-1029, 2017 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-28762673

RESUMEN

BACKGROUND: Neural tube defects (NTDs) are caused by the failure of neural tube formation which occurs during early embryonic development. NTDs are the most severe and leading cause of fetal mortality. Serine hydroxymethyl transferase (SHMT1) provides one-carbon units necessary for embryogenesis and defects in one-carbon production result in specific pathological conditions during pregnancy. The present study is aimed to evaluate the association of SHMT1 C1420T with NTD risk in the fetus using fetal, maternal and paternal groups by applying both case-control and family-based triad approaches. METHODS: A total of 924 subjects including 124 NTD case-parent trios (n = 124 × 3 = 372) and 184 healthy control-parent trios (n = 184 × 3 = 552) from Telangana State, South India were analyzed. DNA from umbilical cord tissues and parental blood samples were extracted, and genotyped by polymerase chain reaction-restriction fragment length polymorphism. Statistical analysis used were SPSS, parent-of-origin effect (POE) analysis. RESULTS: Case-control study design demonstrated fetuses with homozygous variant genotype (TT) to be at risk toward spina bifida subtype (p = 0.022). Among parents, fathers with TT genotype were associated with anencephaly (p = 0.018) and spina bifida subtypes (p = 0.027) in the offspring. Of interest, maternal-paternal-offspring genotype incompatibility revealed maternal CT genotype in combination with paternal TT genotype increased risk for NTDs in the fetus (CTxTT = TT; p = 0.021). Family-based parent-of-origin effect linkage analysis revealed significant maternal over-transmission of variant allele to NTD fetuses (p < 0.01). CONCLUSION: The present study, using both case-control and family-based triad approach is the first report to demonstrate parental association of SHMT1 C1420T variant in conferring NTD risk in the fetus. Birth Defects Research 109:1020-1029, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Glicina Hidroximetiltransferasa/genética , Defectos del Tubo Neural/genética , Adulto , Estudios de Casos y Controles , Susceptibilidad a Enfermedades/complicaciones , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Glicina Hidroximetiltransferasa/metabolismo , Humanos , India , Masculino , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/fisiopatología , Linaje , Polimorfismo de Nucleótido Simple , Embarazo
9.
Neurosci Lett ; 594: 6-11, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25818329

RESUMEN

Neural tube defects (NTDs) are common congenital malformations. Mitogen-activated protein kinases (MAPKs) pathway is involved in many physiological processes. HMGB1 has been showed closely associated with neurulation and NTDs induced by hyperthermia and could activate MAPKs pathway. Since hyperthermia caused increased activation of MAPKs in many systems, the present study aims to investigate whether HMGB1 contributes to hyperthermia induced NTDs through MAPKs pathway. The mRNA levels of MAPKs and HMGB1 between embryonic day 8.5 and 10 (E8.5-10) in hyperthermia induced defective neural tube were detected by real-time quantitative polymerase chain reaction (qPCR). By immunofluorescence and western blotting, the expressions of HMGB1 and phosphorylated MAPKs (ERK1/2, JNK and p38) in neural tubes after hyperthermia were studied. The mRNA levels of MAPKs and HMGB1, as well as the expressions of HMGB1 along with phosphorylated JNK, p38 and ERK, were downregulated in NTDs groups induced by hyperthermia compared with control. The findings suggested that HMGB1 may contribute to hyperthermia induced NTDs formation through decreased cell proliferation due to inhibited phosphorylated ERK1/2 MAPK.


Asunto(s)
Hipertermia Inducida , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Defectos del Tubo Neural/enzimología , Animales , Regulación hacia Abajo , Proteína HMGB1/metabolismo , Sistema de Señalización de MAP Quinasas , Mesocricetus , Proteínas Quinasas Activadas por Mitógenos/genética , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/etiología , Fosforilación , ARN Mensajero/metabolismo
10.
Nat Commun ; 6: 6388, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25736695

RESUMEN

Glycine decarboxylase (GLDC) acts in the glycine cleavage system to decarboxylate glycine and transfer a one-carbon unit into folate one-carbon metabolism. GLDC mutations cause a rare recessive disease non-ketotic hyperglycinemia (NKH). Mutations have also been identified in patients with neural tube defects (NTDs); however, the relationship between NKH and NTDs is unclear. We show that reduced expression of Gldc in mice suppresses glycine cleavage system activity and causes two distinct disease phenotypes. Mutant embryos develop partially penetrant NTDs while surviving mice exhibit post-natal features of NKH including glycine accumulation, early lethality and hydrocephalus. In addition to elevated glycine, Gldc disruption also results in abnormal tissue folate profiles, with depletion of one-carbon-carrying folates, as well as growth retardation and reduced cellular proliferation. Formate treatment normalizes the folate profile, restores embryonic growth and prevents NTDs, suggesting that Gldc deficiency causes NTDs through limiting supply of one-carbon units from mitochondrial folate metabolism.


Asunto(s)
Glicina-Deshidrogenasa (Descarboxilante)/deficiencia , Hiperglicinemia no Cetósica/enzimología , Hiperglicinemia no Cetósica/etiología , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/etiología , Animales , Secuencia de Bases , Cartilla de ADN/genética , Ácido Fólico/metabolismo , Formiatos/farmacología , Galactósidos , Cromatografía de Gases y Espectrometría de Masas , Genotipo , Glicina/metabolismo , Inmunohistoquímica , Hibridación in Situ , Indoles , Ratones , Datos de Secuencia Molecular , Defectos del Tubo Neural/prevención & control , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
11.
Toxicology ; 328: 142-51, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25527867

RESUMEN

Exposure to environmental toxic chemicals in utero during the neural tube development period can cause developmental disorders. To evaluate the disruption of neural tube development programming, the murine neural tube defects (NTDs) model was induced by interrupting folate metabolism using methotrexate in our previous study. The present study aimed to examine the effects of dNTP deficiency induced by hydroxyurea (HU), a specific ribonucleotide reductase (RNR) inhibitor, during murine neural tube development. Pregnant C57BL/6J mice were intraperitoneally injected with various doses of HU on gestation day (GD) 7.5, and the embryos were checked on GD 11.5. RNR activity and deoxynucleoside triphosphate (dNTP) levels were measured in the optimal dose. Additionally, DNA damage was examined by comet analysis and terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL) assay. Cellular behaviors in NTDs embryos were evaluated with phosphorylation of histone H3 (PH-3) and caspase-3 using immunohistochemistry and western blot analysis. The results showed that NTDs were observed mostly with HU treatment at an optimal dose of 225 mg/kg b/w. RNR activity was inhibited and dNTP levels were decreased in HU-treated embryos with NTDs. Additionally, increased DNA damage, decreased proliferation, and increased caspase-3 were significant in NTDs embryos compared to the controls. Results indicated that HU induced murine NTDs model by disturbing dNTP metabolism and further led to the abnormal cell balance between proliferation and apoptosis.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Desoxirribonucleótidos/metabolismo , Inhibidores Enzimáticos/toxicidad , Hidroxiurea/toxicidad , Exposición Materna/efectos adversos , Defectos del Tubo Neural/inducido químicamente , Tubo Neural/efectos de los fármacos , Ribonucleótido Reductasas/antagonistas & inhibidores , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/enzimología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/anomalías , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Daño del ADN , Regulación hacia Abajo , Femenino , Edad Gestacional , Histonas/metabolismo , Ratones Endogámicos C57BL , Tubo Neural/anomalías , Tubo Neural/enzimología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/enzimología , Fosforilación , Embarazo , Ribonucleótido Reductasas/metabolismo , Factores de Tiempo
12.
PLoS One ; 9(6): e101169, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24977710

RESUMEN

OBJECTIVES: The methylenetetrahydrofolate dehydrogenase (MTHFD1) gene, as one of the key genes involved in the folate pathway, has been reported to play a critical role in the pathogenesis of neural tube defects (NTDs). However, the results of published studies are contradictory and inconclusive. Thus, this meta-analysis aimed to evaluate the effect of the common polymorphism in the MTHFD1 gene, the G1958A (R653Q, dbSNP ID: rs2236225) variant, on the risk of NTDs in all eligible studies. METHODS: Relevant literature published before January 3, 2014 was retrieved from the MEDLINE, EMBASE, Cochrane Library, and CBM databases. Pooled crude odds ratios (ORs) and their corresponding 95% confidence intervals (CIs) were calculated to evaluate the association between the MTHFD1 G1958A polymorphism and NTDs risk. RESULTS: We performed a meta-analysis of nine studies with a total of 4,302 NTDs patients and 4,238 healthy controls. Our results demonstrated a significant correlation between the MTHFD1 G1958A polymorphism and NTDs in an overall meta-analysis. For family-based studies, the study subjects were classified as NTD cases, mothers with NTDs offspring, and fathers with NTDs offspring. We found no association between any of the fathers' genotypes and NTDs, whereas there was a clear excess of the 1958A allele in the mothers of children with NTDs compared with controls individuals. CONCLUSIONS: In summary, our meta-analysis strongly suggests that the MTHFD1 G1958A polymorphism might be associated with maternal risk for NTDs in Caucasian populations. However, the evidence of this association should be interpreted with caution due to the selective nature of publication of genetic association studies.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple/genética , Alelos , Humanos , Antígenos de Histocompatibilidad Menor , Oportunidad Relativa , Factores de Riesgo
13.
Birth Defects Res A Clin Mol Teratol ; 100(1): 22-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24382678

RESUMEN

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.


Asunto(s)
Anencefalia/genética , Catecol O-Metiltransferasa/genética , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple , Disrafia Espinal/genética , Té/efectos adversos , Adulto , Anencefalia/inducido químicamente , Anencefalia/enzimología , Estudios de Casos y Controles , Catecol O-Metiltransferasa/metabolismo , China , Femenino , Feto , Predisposición Genética a la Enfermedad , Humanos , Masculino , Exposición Materna/efectos adversos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/enzimología , Oportunidad Relativa , Polifenoles/toxicidad , Factores de Riesgo , Población Rural , Disrafia Espinal/inducido químicamente , Disrafia Espinal/enzimología
14.
Birth Defects Res A Clin Mol Teratol ; 100(1): 13-21, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24307569

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa/genética , Exposición Materna/efectos adversos , Fase II de la Desintoxicación Metabólica/genética , Defectos del Tubo Neural/genética , Hidrocarburos Policíclicos Aromáticos/metabolismo , Polimorfismo de Nucleótido Simple , Adulto , Arilsulfotransferasa/metabolismo , Estudios de Casos y Controles , China , Femenino , Feto , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Masculino , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/etiología , Hidrocarburos Policíclicos Aromáticos/toxicidad , Factores de Riesgo , Población Rural , Esmog/efectos adversos
15.
Int J Clin Exp Pathol ; 6(12): 2968-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24294386

RESUMEN

OBJECTIVE: This study aimed to investigate the single nucleotide polymorphisms (SNPs) of PKA and neural tube defects (NTDs) in Chinese population. METHOD: A total of 183 NTDs cases and 200 healthy controls were used in this study. 7 selected single nucleotide polymorphism (SNP) sites in the PKA gene were analyzed with MassArray high-throughput DNA analyzer with matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry. A series of statistical methods were carried out to investigate the correlation between the SNPs and the patient susceptibility to NTDs. RESULTS: Statistical analysis showed a significant correlation between the SNP sites rs12132032 in PRKACB and NTDs. The AA genotype, A-allele and dominant AA in rs12132032 significantly increased the incidence of NTDs especially anencephaly (OR=3.87, 95% CI: 1.80-8.34 with genotype; OR=2.08, 95% CI: 1.43-3.04 with allele; OR=3.10, 95% CI: 1.53-6.26 with dominant). The T-allele of rs594631 in PRKACB was correlative with NTDs in male but not in female. CONCLUSIONS: The gene polymorphism loci rs12132032 in PRKACB maybe a potential risk factor for anencephaly in Chinese population from Shanxi, while gender susceptibility may influence the correlation.


Asunto(s)
Pueblo Asiatico/genética , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Defectos del Tubo Neural/genética , Anencefalia/enzimología , Anencefalia/etnología , Anencefalia/genética , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , China/epidemiología , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Edad Gestacional , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Incidencia , Modelos Logísticos , Masculino , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/etnología , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Factores Sexuales , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
Gene ; 520(1): 7-13, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23438943

RESUMEN

Many studies have accessed the association between methionine synthase (MTR) A2756G polymorphism and neural tube defect (NTD). However, the conclusions are inconsistent. Our study aimed to clarify the nature of the genetic risks contributed by this polymorphism for NTD using meta-analysis. We searched electronic literature from the PubMed, EMBASE, and Medline databases, from which 10 articles were selected according to the inclusion criteria. The meta-analysis was conducted in 3 groups, namely, NTD patients, mothers with NTD offspring and fathers with NTD offspring. Pooled odds ratios (ORs) and 95% confidence intervals were used to evaluate the strength of the association and the result was corrected by multiple testing. To sum up, no associations between the MTR A2756G polymorphism and NTD risk were found among the 3 groups in all genetic models. However, as their sample size is not large enough, this result needs further research.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Defectos del Tubo Neural/genética , Polimorfismo Genético , Secuencia de Bases , Intervalos de Confianza , Bases de Datos Genéticas , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Defectos del Tubo Neural/enzimología , Oportunidad Relativa , Linaje
17.
Curr Pharm Des ; 19(14): 2574-93, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23116396

RESUMEN

Methylenetetrahydrofolate reductase (MTHFR) catalyzes the reduction of 5,10-methylenetetrahydofolate (CH2-H4folate) to 5-methyltetrahydrofolate (CH3-H4folate). The enzyme employs a noncovalently-bound flavin adenine dinucleotide (FAD), which accepts reducing equivalents from NAD(P)H and transfers them to CH2-H4folate. The reaction provides the sole source of CH3-H4folate, which is utilized by methionine synthase in the synthesis of methionine from homocysteine. MTHFR plays a key role in folate metabolism and in the homeostasis of homocysteine; mutations in the enzyme lead to hyperhomocyst(e)inemia. A common C677T polymorphism in MTHFR has been associated with an increased risk for the development of cardiovascular disease, Alzheimer's disease, and depression in adults, and of neural tube defects in the fetus. The mutation also confers protection for certain types of cancers. This review presents the current knowledge of the enzyme, its biochemical characterization, and medical significance.


Asunto(s)
Hiperhomocisteinemia , Metilenotetrahidrofolato Reductasa (NADPH2) , Polimorfismo de Nucleótido Simple , Regulación Alostérica , Secuencia de Aminoácidos , Animales , Arabidopsis/enzimología , Enfermedades Cardiovasculares/enzimología , Catálisis , Escherichia coli/enzimología , Homocisteína/metabolismo , Humanos , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/genética , Leishmania major/enzimología , Trastornos Mentales/enzimología , Metilenotetrahidrofolato Reductasa (NADPH2)/química , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Neoplasias/enzimología , Defectos del Tubo Neural/enzimología , Alineación de Secuencia , Tetrahidrofolatos/metabolismo
18.
Genet Mol Res ; 10(4): 2424-9, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-22002135

RESUMEN

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.


Asunto(s)
Cistationina betasintasa/genética , Meningomielocele/genética , Mutagénesis Insercional , Defectos del Tubo Neural/genética , Polimorfismo de Longitud del Fragmento de Restricción , Adulto , Preescolar , Cistationina betasintasa/metabolismo , Femenino , Humanos , India/epidemiología , Lactante , Recién Nacido , Masculino , Meningomielocele/enzimología , Meningomielocele/epidemiología , Defectos del Tubo Neural/enzimología , Reacción en Cadena de la Polimerasa/métodos , Prevalencia , Factores de Riesgo
19.
Birth Defects Res A Clin Mol Teratol ; 91(9): 848-56, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21770021

RESUMEN

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.


Asunto(s)
Homocisteína/sangre , Metilenotetrahidrofolato Reductasa (NADPH2) , Defectos del Tubo Neural , Polimorfismo de Nucleótido Simple , Transcobalaminas , Adulto , Estudios de Casos y Controles , Femenino , Ácido Fólico/genética , Ácido Fólico/metabolismo , Predisposición Genética a la Enfermedad , Humanos , India/epidemiología , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/epidemiología , Defectos del Tubo Neural/genética , Transcobalaminas/genética , Transcobalaminas/metabolismo , Vitamina B 12/genética , Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/enzimología , Deficiencia de Vitamina B 12/epidemiología , Deficiencia de Vitamina B 12/genética
20.
Birth Defects Res A Clin Mol Teratol ; 88(10): 883-94, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20842651

RESUMEN

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.


Asunto(s)
Anomalías Congénitas/enzimología , Sistema Enzimático del Citocromo P-450/metabolismo , Desarrollo Embrionario , Tretinoina/metabolismo , Deficiencia de Vitamina A/enzimología , Animales , Anomalías Congénitas/embriología , Anomalías Congénitas/metabolismo , Extremidades/embriología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/enzimología , Embarazo , Ácido Retinoico 4-Hidroxilasa , Teratógenos/metabolismo , Testículo/embriología , Deficiencia de Vitamina A/embriología , Deficiencia de Vitamina A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...