Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Hum Hered ; 88(1): 29-37, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36944328

RESUMO

INTRODUCTION: We have reported that high total homocysteine and the coexistence of inadequate thyroid hormones in maternal serum increase the risk of fetal neural tube defects (NTDs). Placental iodothyronine deiodinases (DIOs: DIO1, DIO2, and DIO3) play a role in regulating the conversions between different forms of maternal thyroid hormones. This study hypothesized that single nucleotide polymorphisms (SNPs) in placental DIOs genes could be related to NTDs. METHODS: We performed a case-control study from 2007 to 2009 that included pregnant women from Lüliang, Shanxi Province, China. Nine distinct SNPs in DIOs genes were analyzed, and placental samples were obtained from 83 pregnant women with NTD fetuses and 90 pregnant women with normal fetuses. The nine SNPs were analyzed using the Cochran-Armitage test and the Fisher's exact test. RESULTS: There were no statistically significant differences between case and control in the nine SNPs of DIOs (p > 0.05). CONCLUSIONS: The results of this study suggested that SNPs of DIO genes in the placenta among pregnant women have no statistically significant difference between the two groups, suggesting that other factors might be involved in metabolism of maternal thyroid hormone provided to fetuses, such as epigenetic modification of methylation and homocysteinylation and genomic imprinting in the placenta. Further functional studies on placenta samples are necessary.


Assuntos
Defeitos do Tubo Neural , Placenta , Gravidez , Humanos , Feminino , Placenta/metabolismo , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Estudos de Casos e Controles , Prevalência , Hormônios Tireóideos/metabolismo , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , China/epidemiologia
2.
Brain ; 146(8): 3455-3469, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36928982

RESUMO

Neural tube defects are the most severe congenital malformations that result from failure of neural tube closure during early embryonic development, and the underlying molecular mechanisms remain elusive. Retinoic acid, an active derivative of vitamin A, is critical for neural system development, and retinoic acid receptor (RAR) signalling malfunctions have been observed in human neural tube defects. However, retinoic acid-retinoic acid receptor signalling regulation and mechanisms in neural tube defects are not fully understood. The mRNA expression of RARs and retinoid X receptors in the different human neural tube defect phenotypes, including 11 pairs of anencephaly foetuses, 10 pairs of hydrocephalus foetuses and nine pairs of encephalocele foetuses, was investigated by NanoString nCounter technology. Immunoprecipitation-mass spectrometry was performed to screen the potential interacting targets of retinoic acid receptor γ. The interactions between proteins were confirmed by co-immunoprecipitation and immunofluorescence laser confocal microscopy. Luciferase and chromatin immunoprecipitation with quantitative real-time polymerase chain reaction assays were used to clarify the underlying mechanism. Moreover, a neural tube defect animal model, constructed using excess retinoic acid, was used for further analysis with established molecular biology technologies. We report that level of retinoic acid receptor γ (RARγ) mRNA was significantly upregulated in the brain tissues of human foetuses with anencephaly. To further understand the actions of retinoic acid receptor γ in neural tube defects, methylenetetrahydrofolate dehydrogenase 1 was identified as a specific retinoic acid receptor γ target from IP-MS screening. Additionally, methylenetetrahydrofolate dehydrogenase 1 negatively regulated retinoic acid receptor γ transcription factor activity. Furthermore, low expression of methylenetetrahydrofolate dehydrogenase 1 and activation of retinoic acid receptor signalling were further determined in human anencephaly and a retinoic acid-induced neural tube defect mouse model. This study reveals that methylenetetrahydrofolate dehydrogenase 1, the rate-determining enzyme in the one-carbon cycle, might be a specific regulator of retinoic acid receptors; these findings provide new insights into the functional linkage between nuclear folate metabolism and retinoic acid receptor signalling in neural tube defect pathology.


Assuntos
Anencefalia , Defeitos do Tubo Neural , Camundongos , Gravidez , Animais , Feminino , Humanos , Metilenotetra-Hidrofolato Desidrogenase (NADP)/efeitos adversos , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Tretinoína/efeitos adversos , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , RNA Mensageiro , Antígenos de Histocompatibilidade Menor
3.
J Trace Elem Med Biol ; 68: 126814, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34243025

RESUMO

BACKGROUND: This study tested the hypothesis that abnormal maternal metabolism of both homocysteine and thyroid hormone network in pregnant women is associated with neural tube defects (NTDs) in a part of China with high NTD prevalence. METHODS: A case-control study was performed between 2007 and 2009 in Lüliang Mountains, Shanxi Province. This study included 83 pregnant women who had fetuses with NTDs (cases) and 90 pregnant women with normal fetuses (controls). In addition, a cell model to illustrate the epidemiological findings was established. RESULTS: Fetuses of mother who had both high total homocysteine (tHcy) and inadequate free thyroxine were 3 times more at risk of developing NTDs (adjusted odds ratio = 3.5; 95 % confidence interval = 1.2-10.4; cases vs. controls) using multivariate logistic regression models. Furthermore, biological interaction between metabolisms of Hcy and thyroid hormones was demonstrated in vitro. In homocysteine thiolactone of a metabolite of Hcy-treated mouse embryonic neural stem NE4C cells, genes (Bmp7, Ctnnb1, Notch 1, Gli2, and Rxra) related to both neural tube closure and thyroid hormone network were shown to be regulated by H3K79 homocysteinylation, which increased their expression levels. CONCLUSIONS: The effect of maternal serum high tHcy on risk of developing NTDs is depended on maternal serum level of thyroxine. Meanwhile, a higher level of tHcy might also affect both maternal metabolism of thyroid hormone and neural tube closure in embryogenesis through homocysteinylation of histones.


Assuntos
Defeitos do Tubo Neural , Gestantes , Animais , Estudos de Casos e Controles , Feminino , Ácido Fólico , Homocisteína , Humanos , Camundongos , Gravidez , Hormônios Tireóideos , Tiroxina
4.
Cell Death Dis ; 12(3): 234, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33664222

RESUMO

Wnt signaling plays a major role in early neural development. An aberrant activation in Wnt/ß-catenin pathway causes defective anteroposterior patterning, which results in neural tube closure defects (NTDs). Changes in folate metabolism may participate in early embryo fate determination. We have identified that folate deficiency activated Wnt/ß-catenin pathway by upregulating a chorion-specific transcription factor Gcm1. Specifically, folate deficiency promoted formation of the Gcm1/ß-catenin/T-cell factor (TCF4) complex formation to regulate the Wnt targeted gene transactivation through Wnt-responsive elements. Moreover, the transcription factor Nanog upregulated Gcm1 transcription in mESCs under folate deficiency. Lastly, in NTDs mouse models and low-folate NTDs human brain samples, Gcm1 and Wnt/ß-catenin targeted genes related to neural tube closure are specifically overexpressed. These results indicated that low-folate level promoted Wnt/ß-catenin signaling via activating Gcm1, and thus leaded into aberrant vertebrate neural development.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Deficiência de Ácido Fólico/metabolismo , Defeitos do Tubo Neural/metabolismo , Tubo Neural/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Deficiência de Ácido Fólico/complicações , Deficiência de Ácido Fólico/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Tubo Neural/anormalidades , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/genética , Gravidez , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Fatores de Transcrição/genética
5.
Epigenomics ; 12(1): 5-18, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31769301

RESUMO

Aim: To know the cause of sequence variants in neural tube defect (NTD). Materials & methods: We sequenced genes implicated in neural tube closure (NTC) in a Chinese cohort and elucidated the molecular mechanism-driving mutations. Results: In NTD cases, an increase in specific variants was identified, potentially deleterious rare variants harbored in H3K36me3 occupancy regions that recruits mismatch repair (MMR) machinery. Lower folate concentrations in local brain tissues were also observed. In neuroectoderm cells, folic acid insufficiency attenuated association of Msh6 to H3K36me3, and reduced bindings to NTC genes. Rare variants in human NTDs were featured by MMR deficiency and more severe microsatellite instability. Conclusion: Our work suggests a mechanistic link between folate insufficiency and MMR deficiency that correlates with an increase of rare variants in NTC genes.


Assuntos
Reparo de Erro de Pareamento de DNA/genética , Suscetibilidade a Doenças , Deficiência de Ácido Fólico/complicações , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/metabolismo , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/metabolismo , Variação Genética , Histonas/metabolismo , Humanos
6.
FASEB J ; 33(4): 4688-4702, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30592646

RESUMO

Folate deficiency in early development leads to disturbance in multiple processes, including neurogenesis during which fibroblast growth factor (FGF) pathway is one of the crucial pathways. Whether folic acid (FA) directly affects FGF pathways to influence neurodevelopment and the possible mechanism remains unclear. In this study, we presented evidence that in human FA-insufficient encephalocele, the FGF pathway was interfered. Furthermore, in Brachyury knockout mice devoid of such T-box transcription factors regulating embryonic neuromesodermal bipotency and a key component of FGF pathway, change in expression of Brachyury downstream targets, activator Fgf8 and suppressor dual specificity phosphatase 6 was detected, along with the reduction in expression of other key FGF pathway genes. By using a FA-deficient cell model, we further demonstrated that decrease in Brachyury expression was through alteration in hypermethylation at the Brachyury promoter region under FA deficiency conditions, and suppression of Brachyury promoted the inactivation of the FGF pathway. Correspondingly, FA supplementation partially reverses the effects seen in FA-deficient embryoid bodies. Lastly, in mice with maternal folate-deficient diets, aberrant FGF pathway activity was found in fetal brain dysplasia. Taken together, our findings highlight the effect of FA on FGF pathways during neurogenesis, and the mechanism may be due to the low expression of Brachyury gene via hypermethylation under FA-insufficient conditions.-Chang, S., Lu, X., Wang, S., Wang, Z., Huo, J., Huang, J., Shangguan, S., Li, S., Zou, J., Bao, Y., Guo, J., Wang, F., Niu, B., Zhang, T., Qiu, Z., Wu, J., Wang, L. The effect of folic acid deficiency on FGF pathway via Brachyury regulation in neural tube defects.


Assuntos
Proteínas Fetais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico , Defeitos do Tubo Neural/tratamento farmacológico , Defeitos do Tubo Neural/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Encefalocele/metabolismo , Feminino , Deficiência de Ácido Fólico/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Sulfitos/farmacologia
7.
BMC Med Genomics ; 11(1): 38, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618362

RESUMO

BACKGROUND: Mouse homozygous mutants in Wnt/planar cell polarity (PCP) pathway genes have been shown to cause neural tube defects (NTDs) through the disruption of normal morphogenetic processes critical to neural tube closure (NTC). Knockout mice that are heterozygotes of single PCP genes likely fail to produce NTD phenotypes, yet damaging variants detected in human NTDs are almost always heterozygous, suggesting that other deleterious interacting variants are likely to be present. Nonetheless, the Wnt/PCP pathway remains a genetic hotspot. Addressing these issues is essential for understanding the genetic etiology of human NTDs. METHODS: We performed targeted next-generation sequencing (NGS) on 30 NTD-predisposing Wnt/PCP pathway genes in 184 Chinese NTD cases. We subsequently replicated our findings for the CELSR1 gene in an independent cohort of 292 Caucasian NTD samples from the USA. Functional validations were confirmed using in vitro assays. RESULTS: CELSR1, CELSR2 and CELSR3 genes were significantly clustered with rare driver coding mutations (q-value< 0.05) demonstrated by OncodriveCLUST. During the validation stage, the number of rare loss of function (LoF) variants in CELSR1 was significantly enriched in NTDs compared with the LoF counts in the ExAC database (p < 0.001). Functional studies indicated compound heterozygote variants of CELSR2 p.Thr2026Met and DVL3 p.Asp403Asn result in down regulation of PCP signals. CONCLUSIONS: These data indicate rare damaging variants of the CELSR genes, identified in ~ 14% of NTD cases, are expected to be driver genes in the Wnt/PCP pathway. Compound damaging variants of CELSR genes and other Wnt/PCP genes, which were observed in 3.3% of the studied NTD cohort, are also expected to amplify these effects at the pathway level.


Assuntos
Polaridade Celular/genética , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/patologia , Proteínas Wnt/genética , Caderinas/genética , Criança , Predisposição Genética para Doença , Células HEK293 , Células HeLa , Humanos , Mutação
8.
CNS Neurosci Ther ; 24(5): 394-403, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29365368

RESUMO

AIM: Neural tube defects (NTDs) are birth defects of the nervous system and are the second most frequent cause of birth defects worldwide. The etiology of NTDs is complicated and involves both genetic and environmental factors. CASP9 is an initiator caspase in the intrinsic apoptosis pathway, which in Casp9-/- mice has been shown to result in NTDs because of decreased apoptosis. The aim of this study was to evaluate the potential genetic contribution of the CASP9 gene in human NTDs. METHODS: High-throughput sequencing was performed to screen genetic variants of CASP9 genes in 355 NTD cases and 225 matched controls. Apoptosis-relevant assays were performed on transiently transfected E9 neuroepithelial cells or human embryonic kidney 293T cells, to determine the functional characteristics of NTD-specific rare variants under complete or low folic acid (FA) status. RESULTS: We found significant expression of CASP9 rare variants in NTDs and identified 4 NTD-specific missense variants. Functional assays demonstrated that a p.Y251C variant attenuates apoptosis by reducing CASP9 protein expression and decreasing activity of the intrinsic apoptosis pathway. From this, we conclude that this variant may represent a loss-of-function mutation. A 4-time recurrent p.R191G variant did not affect intrinsic apoptosis in complete medium, while it completely inhibited apoptosis induced by low FA medium. CONCLUSION: Our findings identify a genetic link for apoptosis in human NTDs and highlight the effect of gene-environment interactions in a complex disease.


Assuntos
Caspase 9/genética , Caspase 9/metabolismo , Mutação , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Apoptose/fisiologia , Povo Asiático/genética , Linhagem Celular , China , Estudos de Coortes , Feminino , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/metabolismo , Expressão Gênica , Interação Gene-Ambiente , Testes Genéticos , Humanos , Masculino
9.
Cell Mol Neurobiol ; 38(3): 605-614, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28623428

RESUMO

Although more than 200 genes are known to be related to neural tube defects (NTDs), the exact molecular basis is still unclear. Evaluating the contribution of copy number variation (CNV) might be a priority because CNV involves changes in the copy number of large segments of DNA, leading to phenotypic traits and disease susceptibility. Recent studies have documented that the polarity protein partitioning defective 3 homolog (Pard3) plays an essential role in the process of neural tube closure. The aim of this study was to assess the role of PARD3 CNVs in the etiology of human NTDs. Relative quantitative PCR and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry were used to quantitative measurement of CNVs in 25 PARD3 exons in 202 NTD cases and 231 controls from a region of China with a high prevalence of NTDs. The results showed that microduplications ranging from 3 to 4 were evident in coding Exon 21 and Exon 25 in both case and control groups. A novel heterozygous microdeletion spanning 444 bp of Exon 14 was identified in two cases of anencephaly and is absent from all controls analyzed. Expression analyses indicated that this heterozygotic microdeletion showed no tissue specificity and led to defective expression of PARD3. Our study provides further evidence implicating PARD3 in the etiology of NTDs.


Assuntos
Proteínas de Ciclo Celular/genética , Variações do Número de Cópias de DNA/genética , Predisposição Genética para Doença/genética , Proteínas de Membrana/genética , Defeitos do Tubo Neural/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Proteínas de Ciclo Celular/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real/métodos
10.
Hum Mutat ; 38(4): 378-389, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27925688

RESUMO

Increasing evidence that mutation of planar cell polarity (PCP) genes contributes to human cranial neural tube defect (NTD) susceptibility prompted us to hypothesize that rare variants of genes in the core apical-basal polarity (ABP) pathway are risk factors for cranial NTDs. In this study, we screened for rare genomic variation of PARD3 in 138 cranial NTD cases and 274 controls. Overall, the rare deleterious variants of PARD3 were significantly associated with increased risk for cranial NTDs (11/138 vs.7/274, P < 0.05, OR = 3.3). These NTD-specific variants were significantly enriched in the aPKC-binding region (6/138 vs. 0/274, P < 0.01). The East Asian cohort in the ExAC database and another Chinese normal cohort further supported this association. Over-expression analysis in HEK293T and MDCK cells confirmed abnormal aPKC binding or interaction for two PARD3 variants (p.P913Q and p.D783G), resulting in defective tight junction formation via disrupted aPKC binding. Functional analysis in human neural progenitor cells and chick embryos revealed that PARD3 knockdown gave rise to abnormal cell polarity and compromised the polarization process of neuroepithelial tissue. Our studies suggest that rare deleterious variants of PARD3 in the aPKC-binding region contribute to human cranial NTDs, possibly by disrupting apical tight junction formation and subsequent polarization process of the neuroepithelium.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Membrana/genética , Mutação , Defeitos do Tubo Neural/genética , Proteína Quinase C/metabolismo , Junções Íntimas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Povo Asiático/genética , Padronização Corporal/genética , Proteínas de Ciclo Celular/metabolismo , Embrião de Galinha , China , Estudos de Coortes , Cães , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Proteínas de Membrana/metabolismo , Defeitos do Tubo Neural/etnologia , Defeitos do Tubo Neural/metabolismo , Ligação Proteica , Interferência de RNA , Junções Íntimas/patologia
11.
G3 (Bethesda) ; 6(10): 3307-3316, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-27543293

RESUMO

Neural tube defects (NTDs) are severe malformations of the central nervous system caused by complex genetic and environmental factors. Among genes involved in NTD, cilia-related genes have been well defined and found to be essential for the completion of neural tube closure (NTC). We have carried out next-generation sequencing on target genes in 373 NTDs and 222 healthy controls, and discovered eight disease-specific rare mutations in cilia-related gene DNAAF1 DNAAF1 plays a central role in cytoplasmic preassembly of distinct dynein-arm complexes, and is expressed in some key tissues involved in neural system development, such as neural tube, floor plate, embryonic node, and brain ependyma epithelial cells in zebrafish and mouse. Therefore, we evaluated the expression and functions of mutations in DNAAF1 in transfected cells to analyze the potential correlation of these mutants to NTDs in humans. One rare frameshift mutation (p.Gln341Argfs*10) resulted in significantly diminished DNAAF1 protein expression, compared to the wild type. Another mutation, p.Lys231Gln, disrupted cytoplasmic preassembly of the dynein-arm complexes in cellular assay. Furthermore, results from NanoString assay on mRNA from NTD samples indicated that DNAAF1 mutants altered the expression level of NTC-related genes. Altogether, these findings suggest that the rare mutations in DNAAF1 may contribute to the susceptibility for NTDs in humans.


Assuntos
Cílios/genética , Estudos de Associação Genética , Mutação , Defeitos do Tubo Neural/genética , Adolescente , Adulto , Alelos , Sequência de Aminoácidos , Substituição de Aminoácidos , Estudos de Casos e Controles , Linhagem Celular , Análise Mutacional de DNA , Feminino , Expressão Gênica , Variação Genética , Genótipo , Humanos , Masculino , Defeitos do Tubo Neural/diagnóstico , Neurogênese/genética , Fenótipo , Adulto Jovem
12.
Birth Defects Res A Clin Mol Teratol ; 106(4): 232-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27001897

RESUMO

BACKGROUND: The polymorphism of genes involved in folate-mediated one-carbon metabolism may be a risk factor for neural tube defects (NTDs). In the present study, we aimed to investigate the single nucleotide polymorphisms (SNPs) of the genes BHMT, CUBN, FTCD, GAMT, GART, SARDH, SHMT1, and MUT, and their effect on NTDs in the Chinese Han population. METHODS: A total of 270 NTDs cases and 192 controls were enrolled in this study. The SNPs were analyzed with the next-generation sequencing method. The folate levels of brain tissues from 113 available NTDs cases and 123 available controls were measured. RESULTS: Next-generation sequencing identified 818 single nucleotide variants, including 214 SNPs used for further analysis. Statistical analysis showed that two independent SNP loci, rs2797840 and rs2073817 in SARDH, may be associated with the susceptibility of NTDs. Specifically, the minor allele G of rs2797840 was significantly associated with NTDs risk in spina bifida subgroup (p value = 0.0348). For subjects whose folate content was measured, the protective allele G of rs2797840 was significantly associated with increased folate content of brain. rs2797840 is within several ENCODE regulatory regions, indicating this SNPs may influence expression of SARDH. CONCLUSION: The SNPs rs2797840 and rs2073817 in SARDH may serve as an indicator for the occurrence of NTDs in the Chinese Han population, and rs2797840 may also be an indicator for folate content of brain.


Assuntos
Ácido Fólico/genética , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único , Sarcosina Desidrogenase/genética , China , Feminino , Ácido Fólico/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Defeitos do Tubo Neural/metabolismo , Sarcosina Desidrogenase/metabolismo
13.
Neurol Sci ; 37(7): 1049-54, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26993122

RESUMO

We investigated single-nucleotide polymorphisms (SNPs) in the aldehyde dehydrogenase family1 L1 gene (ALDH1L1) and their association with neural tube defects (NTDs) in the Chinese population. A total of 271 NTDs cases and 192 healthy controls were used in this study. A total of 112 selected SNPs in the ALDH1L1 gene were analyzed using the next-generation sequencing method. Statistical analysis was carried out to investigate the correlation between SNPs and patient susceptibility to NTDs. Statistical analysis revealed a significant correlation between the SNP sites rs4646733, rs2305225, and rs2276731 in the ALDH1L1 gene and NTDs. The TT genotype and T allele of rs4646733 in ALDH1L1 were associated with a significantly increased incidence of NTDs [odds ratio (OR) = 2.16, 95 % confidence interval (CI) 1.199-3.896 for genotype; and OR = 1.46, 95 % CI 1.092-1.971 for allele]. The AA genotype and A allele of rs2305225 in ALDH1L1 were associated with a significantly increased incidence of NTDs (OR = 2.03, 95 % CI 1.202-3.646 for genotype, and OR = 1.44, 95 % CI 1.096-1.905 for allele). The CT genotype and C allele of rs2276731 in ALDH1L1 significantly were associated with an increased incidence of NTDs (OR = 1.67, 95 % CI 1.129-2.491 with genotype, and OR = 1.32, 95 % CI 0.956-1.816 with allele).The polymorphic loci rs4646733, rs2305225, and rs2276731 in the ALDH1L1 gene maybe potential risk factors for NTDs in the Chinese population.


Assuntos
Aldeído Desidrogenase/genética , Predisposição Genética para Doença/etnologia , Predisposição Genética para Doença/genética , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único/genética , Alelos , Povo Asiático/etnologia , Povo Asiático/genética , Feminino , Estudos de Associação Genética , Genótipo , Humanos , Masculino , Oxirredutases atuantes sobre Doadores de Grupo CH-NH , Fatores de Risco
14.
Mol Neurobiol ; 53(8): 5413-24, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26446020

RESUMO

GLI2 is a key mediator of the sonic hedgehog (Shh) signaling pathway and plays an important role in neural tube development during vertebrate embryogenesis; however, the role of gli2 in human folate-related neural tube defects remains unclear. In this study, we compared methylation status and polymorphisms of gli2 between spina bifida patients and a control group to explore the underlying mechanisms related to folate deficiency in spina bifida. No single nucleotide polymorphism was found to be significantly different between the two groups, although gli2 methylation levels were significantly increased in spina bifida samples, accompanied by aberrant GLI2 expression. Moreover, a prominent negative correlation was found between the folate level in brain tissue and the gli2 methylation status (r = -0.41, P = 0.014), and gli2 hypermethylation increased the risk of spina bifida with an odds ratio of 12.45 (95 % confidence interval: 2.71-57.22, P = 0.001). In addition, we established a cell model to illustrate the effect of gli2 expression and the accessibility of chromatin affected by methylation. High gli2 and gli1 mRNA expression was detected in 5-Aza-treated cells, while gli2 hypermethylation resulted in chromatin inaccessibility and a reduced association with nuclear proteins containing transcriptional factors. More meaningful to the pathway, the effect gene of the Shh pathway, gli1, was found to have a reduced level of expression along with a decreased expression of gli2 in our cell model. Aberrant high methylation resulted in the low expression of gli2 in spina bifida, which was affected by the change in chromatin status and the capacity of transcription factor binding.


Assuntos
Metilação de DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Transdução de Sinais , Disrafismo Espinal/genética , Proteína Gli2 com Dedos de Zinco/genética , Adulto , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Cromatina/metabolismo , Feminino , Ácido Fólico/metabolismo , Humanos , Masculino , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Transcrição Gênica , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo
15.
Med Sci Monit ; 21: 2630-7, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26343515

RESUMO

BACKGROUND: The polymorphism of methylenetetrahydrofolate dehydrogenase (MTHFD1) has been reported as a risk factor for neural tube defects (NTDs). In the present study, we aimed to investigate whether the single-nucleotide polymorphisms (SNPs) of MTHFD1 gene are associated with NTDs in a Chinese population and to determine their mechanism of action. MATERIAL AND METHODS: MTHFD1 gene was scanned in a total of 270 NTDs cases and 192 healthy controls by using next-generation sequencing (NGS) method. After quality control procedures, 208 selected SNP sites in MTHFD1 gene were enrolled for follow-up statistical association analyses. Functional analyses were also performed for significant SNPs through bioinformatics analysis. Folic acid levels of brain tissue in available NTDs cases and healthy controls (113 and 123, respectively) were measured. Statistical and bioinformatics analyses were performed to investigate the relationship between SNPs in MTHFD1 and susceptibility to NTDs. RESULTS: Statistical analysis showed that 2 independent SNPs, rs1956545 and rs56811449, confer the risk of NTDs (P value=0.0195, OR (odds ratio)=1.41, 95% CI (confidence interval)=1.06-1.88; P value=0.0107, OR=0.56, 95% CI=0.36-0.87). The haplotype GGGG, which consists of 4 SNPs (rs2236225, rs2236224, rs1256146, and rs6573559), is also associated with risk of NTDs (P value=0.0438, OR=0.7180, 95% CI=0.5214-0.9888). The risk allele C of rs1956545 is also associated with decreased folic acid levels in the brain (P value=0.0222, standard beta=-0.2238, 95% CI=-0.4128 - -0.0349) according to analysis in the subset of NTDs cases and healthy controls. Bioinformatics analysis indicates that rs1956545 and rs56811449 are within ENCODE regulatory regions, the open chromatin regions of blastula Trophoblast cell line, and histone-marked region of brain astrocyte cell line. CONCLUSIONS: The polymorphism of SNP loci rs1956545 and rs56811449 as well as a haplotype in MTHFD1 gene could serve as an indicator for the occurrence of NTDs in Chinese population and some specific genotypes of the loci may have lower risk of developing NTDs.


Assuntos
Predisposição Genética para Doença , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único , Alelos , Encéfalo/metabolismo , Estudos de Casos e Controles , China , Biologia Computacional/métodos , Feto/patologia , Ácido Fólico/química , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Antígenos de Histocompatibilidade Menor , Defeitos do Tubo Neural/etnologia , Razão de Chances , Polimorfismo Genético , Controle de Qualidade , Fatores de Risco , Natimorto
16.
Gene ; 573(2): 254-60, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26188161

RESUMO

Previous studies have highlighted the connections between neural tube defects (NTDs) and both thyroid hormones (TH) and vitamin A. However, whether the two hormonal signaling pathways interact in NTDs has remained unclear. We measured the expression levels of TH signaling genes in human fetuses with spinal NTDs associated with maternal hyperthyroidism as well as levels of retinoic acid (RA) signaling genes in mouse fetuses exposed to an overdose of RA using NanoString or real-time PCR on spinal cord tissues. Interactions between the two signaling pathways were detected by ChIP assays. The data revealed attenuated DIO2/DIO3 switching in fetuses with NTDs born to hyperthyroid mothers. The promoters of the RA signaling genes CRABP1 and RARB were ectopically occupied by increased RXRG and RXRB but displayed decreased levels of inhibitory histone modifications, suggesting that elevated TH signaling abnormally stimulates RA signaling genes. Conversely, in the mouse model, the observed decrease in Dio3 expression could be explained by increased levels of inhibitory histone modifications in the Dio3 promoter region, suggesting that overactive RA signaling may ectopically derepress TH signaling. This study thus raises in vivo a possible abnormal cross-promotion between two different hormonal signals through their common RXRs and the subsequent recruitment of histone modifications, prompting further investigation into their involvement in the etiology of spinal NTDs.


Assuntos
Defeitos do Tubo Neural/sangue , Complicações na Gravidez/sangue , Hormônios Tireóideos/sangue , Tretinoína/sangue , Animais , Sequência de Bases , Células Cultivadas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Humanos , Hipertireoidismo/sangue , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Defeitos do Tubo Neural/genética , Gravidez , Complicações na Gravidez/genética , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional
17.
Birth Defects Res A Clin Mol Teratol ; 103(1): 37-44, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25131656

RESUMO

BACKGROUND: Animal models of neural tube defects (NTDs) have indicated roles for the Fzd3 gene and the planar cell polarity signaling pathway in convergent extension. We investigated the involvement of FZD3 in genetic and epigenetic mechanisms associated with human NTDs, especially spina bifida. We explored the effects of variants spanning the FZD3 gene in NTDs and examined the role of aberrant methylation of the FZD3 promoter on gene expression in brain tissue in spina bifida. METHODS: Six FZD3 single nucleotide polymorphisms were genotyped using a MassARRAY system in tissue from 165 NTD fetuses and 152 controls. DNA methylation aberrations in the FZD3 promoter region were detected using a MassARRAY EpiTYPER (17 CpG units from -500 to -2400 bp from the transcription start site) in brain tissue from 77 spina bifida and 74 control fetuses. RESULTS: None of the six single nucleotide polymorphisms evaluated were significantly associated with spina bifida, but the mean methylation level was significantly higher in spina bifida samples (13.70%) compared with control samples (10.91%) (p = 0.001). In terms of specific sites, DNA methylation levels were significantly higher in the spina bifida samples at 14 of the 17 CpG units, which mostly included in R2 region. FZD3 mRNA expression was negatively correlated with methylation of the FZD3 promoter region, especially the R2 region (R = 0.970; p = 0.001) in HeLa cells. CONCLUSION: The results of this study suggest that DNA methylation plays an important role in FZD3 gene expression regulation and may be associated with an increased risk of spina bifida.


Assuntos
Metilação de DNA , Receptores Frizzled/genética , Regulação da Expressão Gênica , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Disrafismo Espinal/etiologia , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Epigênese Genética , Feminino , Feto/metabolismo , Feto/patologia , Predisposição Genética para Doença , Genótipo , Idade Gestacional , Humanos , Masculino , Dados de Sequência Molecular , Defeitos do Tubo Neural/complicações , Gravidez , Fatores de Risco , Disrafismo Espinal/patologia
18.
PLoS One ; 9(11): e113308, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25423083

RESUMO

The methylation status of DNA methylation regions (DMRs) of the imprinted gene IGF2/Igf2 is associated with neural tube defects (NTDs), which are caused by a failure of the neural tube to fold and close and are the second-most common birth defect; however, the characterization of the expression level of IGF2/Igf2 in neural tissue from human fetuses affected with NTDs remains elusive. More importantly, whether abnormal chromatin structure also influences IGF2/Igf2 expression in NTDs is unclear. Here, we investigated the transcriptional activity of IGF2/Igf2 in normal and NTD spinal cord tissues, the methylation status of different DMRs, and the chromatin structure of the promoter. Our data indicated that in NTD samples from both human fetuses and retinoic acid (RA)-treated mouse fetuses, the expression level of IGF2/Igf2 was upregulated 6.41-fold and 1.84-fold, respectively, compared to controls. H19 DMR1, but not IGF2 DMR0, was hypermethylated in human NTD samples. In NTD mice, h19 DMR1 was stable, whereas the chromatin structure around the promoter of Igf2 might be loosened, which was displayed by higher H3K4 acetylation and lower H3K27 trimethylation. Therefore, the data revealed that IGF2/Igf2 expression can be ectopically up-regulated by dual epigenetic factors in NTDs. In detail, the upregulation of IGF2/Igf2 is likely controlled by hypermethylation of H19 DMR1 in human NTDs, however, in acute external RA-induced NTD mice it is potentially determined by more open chromatin structure.


Assuntos
Epigênese Genética , Fator de Crescimento Insulin-Like II/genética , Defeitos do Tubo Neural/genética , Regulação para Cima , Animais , Imunoprecipitação da Cromatina , Metilação de DNA , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Defeitos do Tubo Neural/induzido quimicamente , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica , Tretinoína/toxicidade
19.
Int J Clin Exp Pathol ; 6(12): 2968-74, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24294386

RESUMO

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.


Assuntos
Povo Asiático/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Defeitos do Tubo Neural/genética , Anencefalia/enzimologia , Anencefalia/etnologia , Anencefalia/genética , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , China/epidemiologia , Feminino , Frequência do Gene , Predisposição Genética para Doença , Idade Gestacional , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Incidência , Modelos Logísticos , Masculino , Defeitos do Tubo Neural/enzimologia , Defeitos do Tubo Neural/etnologia , Razão de Chances , Fenótipo , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Fatores Sexuais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
20.
Neurobiol Dis ; 54: 404-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23376398

RESUMO

Neural tube defects (NTDs) are severe, common birth defects that result from failure of neural tube closure, but their pathological mechanisms are not yet fully understood. Histone modifications have an important role in gene regulation during fetal development. We therefore hypothesized that the human NTDs may be partly caused by an imbalance in metabolism, perhaps caused by nutritional deficiencies, that leads to aberrant histone modifications. Here, we report a screen of fetal brain histone modifications using 2D nano-LC strong cation exchange reverse phase (SCX/RP) MS/MS and the identification of 61 unique post-translational modification sites on histones H1, H2a, H2b, H3, and H4. Of these, 38 sites are novel (not already found in the Uniprot database). Furthermore, we compared the histone modification patterns between normal brains and NTD brains special of which maternal folate levels were lower than of normal control. The results showed that histone H3 lysine 79 dimethylation (H3K79me2) and a novel identified site, H2bK5 monomethylation (H2bK5me1), were completely absent in individuals with NTDs. Follow-up Western blotting validated the decreased H3K79me2 expression in brains with NTDs, but the amplified samples experiments displayed that decreased H3K79me2 expression was not suitable for all samples with NTDs. Furthermore, folate-free treated mouse embryonic stem cells induced the decreased H3K79me2 level. Subsequently, our ChIP results in normal fetal brain tissues showed that H3K79me2 binds to SUFU, RARA and ITGA3 which induce NTDs phenotype after knockout in mice, and in NTDs brain tissues the bindings of H3K79me2 to these three genes were significantly altered. Taken together, our study indicated that low folate treatment might attenuate H3K79 dimethylation, further affect its regulate activation on target genes, some of which are NTDs-resulting associated, lastly interrupt early embryo developing. Our study increases the understanding of normal fetal brain histone modifications and provides a platform for investigating histone modifications in neural disease and also has an insight into a potential role of aberrant histone modification in etiology of NTDs.


Assuntos
Histonas/genética , Defeitos do Tubo Neural/genética , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Western Blotting , Imunoprecipitação da Cromatina , Feminino , Feto , Histonas/metabolismo , Humanos , Lisina/genética , Masculino , Metilação , Camundongos , Defeitos do Tubo Neural/metabolismo , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA