Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain ; 146(8): 3455-3469, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36928982

RESUMEN

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.


Asunto(s)
Anencefalia , Defectos del Tubo Neural , Ratones , Embarazo , Animales , Femenino , Humanos , Metilenotetrahidrofolato Deshidrogenasa (NADP)/efectos adversos , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Tretinoina/efectos adversos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , ARN Mensajero , Antígenos de Histocompatibilidad Menor
2.
Pediatr Res ; 92(4): 987-994, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34934172

RESUMEN

BACKGROUND: Paternally expressed gene 10 (PEG10) is believed to be a key imprinted gene involved in placenta formation. However, its role in human folate-related spina bifida (SB) remains unclear. METHODS: The methylation status of the germline differentially methylated region (gDMR) in the PEG10/sarcoglycan epsilon (SGCE) imprinted cluster was compared between SB patients and control samples. Moreover, the influence of ectopic PEG10 expression on apoptosis was assessed to explore the underlying mechanisms related to folate deficiency-induced aberrant gDMR methylation in SB. RESULTS: The case group exhibited a significant increase in the methylation level of the gDMR and a marked reduction in the mRNA and protein expression of PEG10 compared with the control group. A prominent negative correlation was found between the folate level in brain tissue and gDMR methylation status (r = -0.62, P = 0.001). A cell model treated with a demethylating agent showed a significant elevation of PEG10 transcription level, as well as other imprinted genes in this cluster. In addition, the inhibition of PEG10 was found to be accompanied by aberrant activation of apoptosis in SB. CONCLUSIONS: Our findings suggest that disturbed gDMR methylation of the PEG10/SGCE cluster due to folate deficiency is involved in SB through aberrant activation of apoptosis. IMPACT: Disturbed genomic imprinting has been verified to be involved in neural tube defects (NTDs). However, little is known about the effect of ectopic expression of imprinted gene PEG10 on human NTDs. Aberrant methylation status of the germline differentially methylated region (gDMR) of PEG10/SGCE cluster due to folate deficiency has been found to result in the inhibition of PEG10 and has a marked association with an increased occurrence of spina bifida. Inhibited expression of PEG10 partly is found to be related to the abnormal activation of apoptosis in spina bifida.


Asunto(s)
Deficiencia de Ácido Fólico , Defectos del Tubo Neural , Disrafia Espinal , Embarazo , Femenino , Humanos , Metilación de ADN , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Deficiencia de Ácido Fólico/genética , Disrafia Espinal/genética , Ácido Fólico , ARN Mensajero/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ARN/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo
3.
Int J Neurosci ; 131(5): 468-477, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32241207

RESUMEN

OBJECTIVE: Neural tube defects (NTDs) are one of the most common and serious birth defects in human beings caused by genetic and environmental factors. Folate insufficiency is involved in the occurrence of NTDs and folic acid supplementation can prevent NTDs occurrence, however, the underlying mechanism remains poorly understood. METHODS: We established cell and animal models of folic acid deficiency to detect the methylation modification and expression levels of genes by MassARRAY and real-time PCR, respectively. Results and conclusion: In the present study, we found firstly that in human folic acid-insufficient NTDs, the methylation level of imprinted gene Mest/Peg1 was decreased. By using a folic acid-deficient cell model, we demonstrated that Mest/Peg1 methylation was descended. Meanwhile, the mRNA level of Mest/Peg1 was up-regulated via hypomethylation modification under low folic acid conditions. Consistent with the results in cell models, Mest/Peg1 expression was elevated through hypomethylation regulation in folate-deficient animal models. Furthermore, the up-regulation of Mest/Peg1 inhibited the expression of Lrp6 gene, a crucial component of Wnt pathway. Similar results with Lrp6 down-regulation of fetal brain were verified in animal models under folic acid-deficient condition. Taken together, our findings indicated folic acid increased the expression of Mest/Peg1 via hypomethylation modification, and then inhibited Lrp6 expression, which may ultimately impact on the development of nervous system through the inactivation of Wnt pathway.


Asunto(s)
Encéfalo/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Defectos del Tubo Neural/metabolismo , Proteínas/metabolismo , Vía de Señalización Wnt/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Feto , Deficiencia de Ácido Fólico/complicaciones , Regulación de la Expresión Génica , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Defectos del Tubo Neural/etiología
4.
J Cell Mol Med ; 24(17): 9898-9907, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32693431

RESUMEN

Congenital heart disease (CHD) with extracardiac malformations (EM) is the most common multiple malformation, resulting from the interaction between genetic abnormalities and environmental factors. Most studies have attributed the causes of CHD with EM to chromosomal abnormalities. However, multi-system dysplasia is usually caused by both genetic mutations and epigenetic dysregulation. The epigenetic mechanisms underlying the pathogenesis of CHD with EM remain unclear. In this study, we investigated the mechanisms of imprinting alterations, including those of the Small nuclear ribonucleoprotein polypeptide N (SNRPN), PLAG1 like zinc finger 1 (ZAC1) and inositol polyphosphate-5-phosphatase F (INPP5F) genes, in the pathogenesis of CHD with EM. The methylation levels of SNRPN, ZAC1, and INPP5F genes were analysed by the MassARRAY platform in 24 children with CHD with EM and 20 healthy controls. The expression levels of these genes were detected by real-time polymerase chain reaction (PCR). The correlation between methylation regulation and gene expression was confirmed using 5-azacytidine (5-Aza) treated cells. The methylation levels of SNRPN and ZAC1 genes were significantly increased in CHD with EM, while that of INPP5F was decreased. The methylation alterations of these genes were negatively correlated with expression. Risk analysis showed that abnormal hypermethylation of SNRPN and ZAC1 resulted in 5.545 and 7.438 times higher risks of CHD with EM, respectively, and the abnormal hypomethylation of INPP5F was 8.38 times higher than that of the control group. We concluded that abnormally high methylation levels of SNRPN and ZAC1 and decreased levels of INPP5F imply an increased risk of CHD with EM by altering their gene functions. This study provides evidence of imprinted regulation in the pathogenesis of multiple malformations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cardiopatías Congénitas/genética , Inositol Polifosfato 5-Fosfatasas/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Nucleares snRNP/genética , Azacitidina/química , Niño , Preescolar , Metilación de ADN/genética , Epigénesis Genética/genética , Femenino , Expresión Génica/genética , Impresión Genómica/genética , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/patología , Humanos , Lactante , Masculino
5.
FASEB J ; 33(4): 4688-4702, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30592646

RESUMEN

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.


Asunto(s)
Proteínas Fetales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/uso terapéutico , Defectos del Tubo Neural/tratamiento farmacológico , Defectos del Tubo Neural/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Encefalocele/metabolismo , Femenino , Deficiencia de Ácido Fólico/fisiopatología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Transducción de Señal/efectos de los fármacos , Sulfitos/farmacología
6.
Med Sci Monit ; 25: 3354-3365, 2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-31061382

RESUMEN

BACKGROUND Maternal folate deficiency-mediated metabolic disruption is considered to be associated with the risk of intrauterine growth retardation (IUGR), but the exact mechanism remains unclear. The retrotransposon long interspersed nucleotide element-1 (LINE-1), which can induce birth defects via RNA intermediates, plays crucial roles during embryonic development. We investigated potential relationships between maternal folate and DNA methylation, and possible roles of LINE-1 in IUGR. MATERIAL AND METHODS The IUGR model was established by feeding female mice 1 of 3 diets - control diet (CD), folate-deficient diet for 2 weeks (FD2w), and folate-deficient diet for 4 weeks (FD4w) - prior to mating. Maternal serum folate, 5-methyltetrahydrofolate (5-MeTHF), S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH) concentrations and global DNA methylation were assessed by LC/MS/MS method. LINE-1 methylation levels in fetuses were examined by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. LINE-1 expression levels were validated by real-time PCR. RESULTS Maternal folate deficiency caused plasma folate and 5-MeTHF levels to decrease and SAH level to increase in the FD4w group. Compared with the CD group, methylation levels of genomic DNA and LINE-1 decreased significantly in placenta and fetal tissues from the FD4w group. Expression of LINE-1 open reading frame 1 (ORF1) protein was elevated in fetal liver tissues. Furthermore, a strong correlation was found between methylation and disrupted one-carbon metabolism, implying that dietary folate plays important roles during embryogenesis. CONCLUSIONS Maternal dietary folate deficiency impaired one-carbon metabolism, leading to global DNA and LINE-1 hypomethylation, and then increased retrotransposition in fetuses, which can lead to IUGR.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Animales , Metilación de ADN/genética , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Feto/metabolismo , Ácido Fólico/sangre , Ácido Fólico/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Elementos de Nucleótido Esparcido Largo/fisiología , Masculino , Intercambio Materno-Fetal/fisiología , Ratones , Ratones Endogámicos C57BL , Placenta/metabolismo , Embarazo , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Tetrahidrofolatos/metabolismo
7.
J Cell Mol Med ; 21(12): 3244-3253, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28631291

RESUMEN

Folic acid deficiency during pregnancy is believed to be a high-risk factor for neural tube defects (NTDs). Disturbed epigenetic modifications, including miRNA regulation, have been linked to the pathogenesis of NTDs in those with folate deficiency. However, the mechanism by which folic acid-regulated miRNA influences this pathogenesis remains unclear. It is believed that DNA methylation is associated with dysregulated miRNA expression. To clarify this issue, here we measured the methylation changes of 22 miRNAs in 57 human NTD cases to explore whether such changes are involved in miRNA regulation in NTD cases through folate metabolism. In total, eight of the 22 miRNAs tested reduced their methylation modifications in NTD cases, which provide direct evidence of the roles of interactions between DNA methylation and miRNA level in these defects. Among the findings, there was a significant association between folic acid concentration and hsa-let-7 g methylation level in NTD cases. Hypomethylation of hsa-let-7 g increased its own expression level in both NTD cases and cell models, which indicated that hsa-let-7 g methylation directly regulates its own expression. Overexpression of hsa-let-7 g, along with its target genes, disturbed the migration and proliferation of SK-N-SH cells, implying that hsa-let-7 g plays important roles in the prevention of NTDs by folic acid. In summary, our data suggest a relationship between aberrant methylation of hsa-let-7 g and disturbed folate metabolism in NTDs, implying that improvements in nutrition during early pregnancy may prevent such defects, possibly via the donation of methyl groups for miRNAs.


Asunto(s)
Epigénesis Genética , Deficiencia de Ácido Fólico/genética , Ácido Fólico/metabolismo , MicroARNs/genética , Defectos del Tubo Neural/genética , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Metilación de ADN , Femenino , Feto , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Humanos , MicroARNs/metabolismo , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Neuronas/metabolismo , Neuronas/patología , Embarazo
8.
Birth Defects Res A Clin Mol Teratol ; 106(8): 667-74, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27173682

RESUMEN

BACKGROUND: The methylenetetrahydrofolate reductase (MTHFR) polymorphism is a risk factor for neural tube defects. C677T and A1298C MTHFR polymorphisms produce an enzyme with reduced folate-related one carbon metabolism, and this has been associated with aberrant methylation modifications in DNA and protein. METHODS: A meta-analysis was conducted to assess the association between MTHFR C677T/A1298C genotypes and global genomic methylation. RESULTS: Eleven studies met the inclusion criteria. Of these, 10 were performed on C677T MTHFR genotypes and 6 were performed on A1298C MTHFR genotypes. Our results did not indicate any correlation between global methylation and MTHFR A1298C, C677T polymorphisms. CONCLUSION: The results of our study provide evidence to assess the global methylation modification alterations of MTHFR polymorphisms among individuals. However, our data did not found any conceivable proof supporting the hypothesis that common variant of MTHFR A1298C, C677T contributes to methylation modification. Birth Defects Research (Part A) 106:667-674, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Metilación de ADN , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple , Femenino , Expresión Génica , Genotipo , Humanos , Masculino , Defectos del Tubo Neural/diagnóstico , Defectos del Tubo Neural/patología , Oportunidad Relativa , Factores de Riesgo
9.
Birth Defects Res A Clin Mol Teratol ; 103(1): 37-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25131656

RESUMEN

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.


Asunto(s)
Metilación de ADN , Receptores Frizzled/genética , Regulación de la Expresión Génica , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Disrafia Espinal/etiología , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Casos y Controles , Epigénesis Genética , Femenino , Feto/metabolismo , Feto/patología , Predisposición Genética a la Enfermedad , Genotipo , Edad Gestacional , Humanos , Masculino , Datos de Secuencia Molecular , Defectos del Tubo Neural/complicaciones , Embarazo , Factores de Riesgo , Disrafia Espinal/patología
10.
J Cell Biochem ; 114(7): 1549-58, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23297156

RESUMEN

Folate is thought to contribute to health and development by methylation regulation. Long interspersed nucleotide element-1 (LINE-1), which is regulated by methylation modification, plays an important role in sculpting the structure and function of genomes. Some studies have shown that folate concentration is related to LINE-1 methylation. However, the direct association between LINE-1 methylation and folate deficiency remains unclear. To explore whether folate deficiency directly induced LINE-1 hypomethylation and to analyze the relationship between folate concentration and the LINE-1 methylation level, mouse ESCs were treated with various concentrations of folate which was measured by chemiluminescent immunoassay, and the homocysteine content was detected by ELISA. LINE-1 methylation was examined by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry at various time points. Concurrently, cell proliferation and differentiation were observed. The result showed that the intracellular folate decreases under folate-deficient condition, conversely, homocysteine content increased gradually and there was a negatively correlated between them. Folate insufficiency induced LINE-1 hypomethylation at the lowest levels in folate-free group and moderate in folate-deficient group, compared with that in the folate-normal group at day 18. Moreover, LINE-1 methylation level was positively correlated with folate content, and negatively correlated with homocysteine content. At corresponding time points, proliferation and differentiation of mouse ESCs showed no alteration in all groups. Our data indicated that folate deficiency affected the homeostasis of folate-mediated one-carbon metabolism, leading to reduced LINE-1 methylation in mouse ESCs. This study provides preliminary evidence of folate deficiency affecting early embryonic development.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Ácido Fólico/farmacología , Elementos de Nucleótido Esparcido Largo/genética , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Células Madre Embrionarias/citología , Ratones
11.
Neurobiol Dis ; 54: 404-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23376398

RESUMEN

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.


Asunto(s)
Histonas/genética , Defectos del Tubo Neural/genética , Procesamiento Proteico-Postraduccional/fisiología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Feto , Histonas/metabolismo , Humanos , Lisina/genética , Masculino , Metilación , Ratones , Defectos del Tubo Neural/metabolismo , Espectrometría de Masas en Tándem
12.
Mol Cell Biochem ; 380(1-2): 33-42, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23690138

RESUMEN

Neural tube defects (NTDs) are serious congenital malformation of fusion failure of the neural tube during early embryogenesis. DNA methylation disorders have been found in NTD-affected fetuses, and are correlated to the risk of NTDs. The insulin-like growth factor 2 (IGF2) gene, maternally imprinted, has a key role in fetal development. IGF2 transcription is partly controlled by differentially methylated regions (DMRs) 0 and 2. To assess whether disturbed methylation pattern increases the incidence of NTDs, we employed matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) to quantify CpG methylation levels of DMR2 and 0 in fetuses with or without NTDs. We found that the methylation level of IGF2 DMR0 increased significantly in the brain tissues of NTD-affected fetuses. And hypermethylation of DMR0 was associated with an increased risk of NTDs, with an odds ratio of 5.375 (95 % CI: 1.447-19.965; p = 0.007). IGF2 mRNA expression was negatively correlated with the methylation level of DMR0 (R (2) = 0.893; p = 0.000) in HCT15 cells. These results highlights that IGF2 DMR0 hypermethylation is a potential risk factor of NTD, and IGF2 gene is a promising candidate gene to study for a greater understanding of the cause of NTDs.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Factor II del Crecimiento Similar a la Insulina/genética , Defectos del Tubo Neural/genética , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Línea Celular Tumoral , Femenino , Feto/embriología , Feto/metabolismo , Edad Gestacional , Humanos , Pulmón/embriología , Pulmón/metabolismo , Masculino , Datos de Secuencia Molecular , Placenta/embriología , Placenta/metabolismo , Reacción en Cadena de la Polimerasa , Embarazo , Factores de Riesgo , Factores Sexuales , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Birth Defects Res A Clin Mol Teratol ; 97(6): 409-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23761049

RESUMEN

BACKGROUND: SHH signaling pathway plays an important role in the formation of the neural plate and is involved in the regulation of the dorsoventral (DV) axis of the neural tube. Some neural tube defects (NTDs) may be caused through overactivation of the SHH signaling pathway. The PTCH1 gene, encoding a negative regulator of SHH signaling, affects neural tube closure in animal models. However, in humans, the relationship between single nucleotide polymorphisms (SNPs) of the PTCH1 gene and neural tube defects remains unclear. METHODS: MassARRAY®GENOTYPER™ was used to genotype 18 SNPs of the PTCH1 gene in 187 NTDs and 212 control samples, to determine whether PTCH1 polymorphisms are related to NTDs. MassARRAY®EpiTYPER™ was performed to assess whether methylation modifications may be associated with SNP genotypes in this Chinese population. RESULT: Increased risk for spina bifida was observed with the G allele of c.3944C>T and the T allele of c.1729™2350G>A in female patients when compared to the normal control group. High methylation levels were detected in those controls bearing the G allele of c.3944C>T. CONCLUSION: In summary, polymorphisms of the PTCH1 gene may be genetic predisposing factors for spina bifida in the population studied. In addition, methylation modifications associated with the c.3944C>T polymorphism, may provide protection.


Asunto(s)
Pueblo Asiatico/genética , Defectos del Tubo Neural/epidemiología , Defectos del Tubo Neural/genética , Receptores de Superficie Celular/genética , Transducción de Señal/fisiología , Metilación de ADN , Femenino , Estudios de Asociación Genética , Genotipo , Proteínas Hedgehog/genética , Humanos , Modelos Logísticos , Receptores Patched , Receptor Patched-1 , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Transducción de Señal/genética
14.
J Nutr Biochem ; 116: 109297, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36907530

RESUMEN

Proper Dlk1-Dio3 imprinting plays a critical role in embryogenesis, and folic acid deficiency may affect the imprinting of this locus through epigenetic regulation. However, whether and how folic acid directly impacts the imprinting status of Dlk1-Dio3 to affect neural development remain unclear. Here, we found decreased IG-DMR (intergenic -differentially methylated regions) methylation in the folate-deficient encephalocele in humans, suggesting that abnormal Dlk1-Dio3 imprinting status is related to neural tube defects (NTDs) caused by folate deficiency. Similar results were obtained with folate-deficient embryonic stem cells. By miRNA chip analysis, folic acid deficiency led to changes in multiple miRNAs, including the upregulation of 15 miRNAs located in the Dlk1-Dio3 locus. Real-time PCR confirmed that seven of these miRNAs were upregulated, especially miR-370. In contrast to normal embryonic development, in which expression of miR-370 is highest at E9.5, the abnormally high and sustained expression of miRNA-370 in folate-deficient E13.5 embryos may contribute to NTDs. In addition, we found that DNMT3A (de novo DNA methyltransferases 3A) is a direct target gene of miR-370 in neural cells, and DNMT3A participates in the role of miR-370 in inhibiting cell migration. Finally, in the folate-deficient mouse model, Dlk1-Dio3 epigenetic activation was found in fetal brain tissue, along with the upregulation of miR-370 and the downregulation of DNMT3A. Collectively, our findings demonstrate a pivotal role of folate in the epigenetic regulation of Dlk1-Dio3 imprinting during neurogenesis, revealing an elegant mechanism for the activation of Dlk1-Dio3 locus miRNAs in folic acid deficiency.


Asunto(s)
Deficiencia de Ácido Fólico , MicroARNs , Humanos , Embarazo , Femenino , Ratones , Animales , Impresión Genómica , Epigénesis Genética , Deficiencia de Ácido Fólico/genética , MicroARNs/genética , MicroARNs/metabolismo , Ácido Fólico , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Metilación de ADN , Proteínas de la Membrana/metabolismo
15.
BMC Med Genomics ; 14(1): 4, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407475

RESUMEN

BACKGROUND: Congenital heart disease (CHD) is resulted from the interaction of genetic aberration and environmental factors. Imprinted genes, which are regulated by epigenetic modifications, are essential for the normal embryonic development. However, the role of imprinted genes in the etiology of CHD remains unclear. METHODS: After the samples were treated with bisulfate salt, imprinted genes methylation were measured by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. T test and One-way ANOVA were performed to evaluate the differences among groups. Odds ratios (ORs) were performed to evaluate the incidence risk of CHD in relation to methylation levels. RESULTS: We investigated the alterations of imprinted gene germline differential methylation regions (gDMRs) methylation in patients with CHD. Eighteen imprinted genes that are known to affect early embryonic development were selected and the methylation modification genes were detected by massarray in 27 CHD children and 28 healthy children. Altered gDMR methylation level of 8 imprinted genes was found, including 2 imprinted genes with hypermethylation of GRB10 and MEST and 6 genes with hypomethylation of PEG10, NAP1L5, INPP5F, PLAGL1, NESP and MEG3. Stratified analysis showed that the methylation degree of imprinted genes was different in different types of CHD. Risk analysis showed that 6 imprinted genes, except MEST and NAP1L5, within a specific methylation level range were the risk factors for CHD CONCLUSION: Altered methylation of imprinted genes is associated with CHD and varies in different types of CHD. Further experiments are warranted to identify the methylation characteristics of imprinted genes in different types of CHD and clarify the etiologies of imprinted genes in CHD.


Asunto(s)
Cardiopatías Congénitas , Niño , Metilación de ADN , Epigénesis Genética , Femenino , Humanos , Masculino , Proyectos Piloto , Embarazo
16.
Mol Genet Genomic Med ; 8(1): e1046, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785132

RESUMEN

BACKGROUND: Human leukocyte antigen (HLA)-DP is an HLA class II molecule. Overexpression of HLA class II molecules in placental trophoblast cells may induce pregnancy loss. However, the association between HLA-DP and pregnancy loss remains unclear. HLA-DPA1 is an HLA-DP peptide chain. The objective of this study was to assess the association between HLA-DPA1 genetic polymorphism and anembryonic pregnancy, a type of early pregnancy loss, in the Chinese population. METHODS: A case-control study was designed to compare the frequencies of HLA-DPA1 gene polymorphisms in an anembryonic pregnancy group and a control group. Sixty-eight cases and 122 controls were recruited. Statistical analysis was performed to assess the correlation between single-nucleotide polymorphisms (SNPs) and anembryonic pregnancy susceptibility. MassARRAY high-throughput DNA analysis was used to analyze 19 HLA-DPA1 SNPs. To explore how HLA-DPA1 polymorphism could affect anembryonic pregnancy, HLA-DPA1 serum levels were analyzed by ELISA. RESULTS: Homozygous typing of rs1431403 (CC and TT) significantly increased the risk of anembryonic pregnancy in the case group (ORCC  = 3.13, 95% CI: 1.50-6.53; ORTT  = 2.96, 95% CI: 1.31-6.66; ORCC+TT  = 3.06, 95% CI: 1.62-5.78). In samples with high HLA-DPA1 levels (≥1,500 pg/ml), the homozygous rs1431403 genotypes (nCC  = 21, 43.8%; nTT  = 20, 57.1%) were observed more frequently than were heterozygous genotypes. CONCLUSION: HLA-DPA1 rs1431403 may be a risk factor for anembryonic pregnancy in the Chinese population. Homozygous rs1431403 genotypes (CC and TT) may increase the risk of anembryonic pregnancy by aberrantly increasing the HLA-DPA1 levels.


Asunto(s)
Aborto Espontáneo/genética , Cadenas alfa de HLA-DP/genética , Polimorfismo de Nucleótido Simple , Aborto Espontáneo/patología , Adulto , Femenino , Humanos , Fenotipo , Embarazo
17.
Epigenomics ; 12(1): 5-18, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769301

RESUMEN

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.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Susceptibilidad a Enfermedades , Deficiencia de Ácido Fólico/complicaciones , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/metabolismo , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Variación Genética , Histonas/metabolismo , Humanos
18.
Int Urol Nephrol ; 50(12): 2193-2200, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30324582

RESUMEN

BACKGROUND: Prostate cancer (PCa) is a common malignant human tumor and one of the main causes of cancer-related deaths in men. At present, prostate-specific antigen levels are widely used to diagnose PCa in the clinic, but they are not sufficient for an accurate early diagnosis or prognosis. METHODS: To identify potential molecular markers for PCa, we used real-time PCR to measure the expression levels of various microRNAs, including miR-1825, miR-484, miR-205, miR-141, and let-7b, in the serum of 72 PCa patients and 34 healthy controls. RESULTS: miR-1825, miR-484, miR-205, miR-141, and let-7b were shown to be highly specific for PCa, suggesting that they could be used as PCa tumor screening biomarkers. miR-205 may also be used as a biomarker for indicating bone metastasis in PCa patients, miR-1825 levels may help indicate tumor-node-metastasis classification, the evaluation of treatment effects, and determining prognosis, while let-7b levels may indicate potential tumor malignancy and the hormone resistance status and could be used as a basis to adjust individual treatments for the high-risk, early diagnosis of refractory PCa. CONCLUSION: This study identified possible PCa tumor markers to more accurately predict the occurrence, progression, and prognosis of PCa, and which could be used in the development of tumor drug therapy.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Óseas/sangre , MicroARNs/sangre , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Anciano , Neoplasias Óseas/diagnóstico , Estudios de Casos y Controles , Humanos , Metástasis Linfática , Masculino , Estadificación de Neoplasias , Pronóstico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/terapia , Curva ROC , Resultado del Tratamiento
19.
Asia Pac J Clin Nutr ; 27(3): 718-727, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29737822

RESUMEN

BACKGROUND AND OBJECTIVES: Methylenetetrahydrofolate reductase (MTHFR) irreversibly converts 5,10- methylenetetrahydrofolate to 5-methyltetrahydrofolate, which is the main form of folate used in the body. Previous studies suggest that MTHFR polymorphism influences folate metabolism, but conflicting results are reported. We performed a meta-analysis to accurately characterize the association between MTHFR A1298C polymorphism and peripheral blood folate concentration in healthy populations. METHODS AND STUDY DESIGN: Studies focusing on MTHFR A1298C polymorphism and folate concentrations were identified and subjected to a metaanalysis using Review Manager 5.1. Standard mean differences (SMD) with 95% confidence intervals (95% CI) were used to assess the association between these variables. RESULTS: A total of 14 studies with 5616 healthy individuals were included in this meta-analysis. Significant differences in folate concentration were found in the MTHFR homozygote model (SMD=0.12, 95% CI=0.00-0.24, I2=17%, p=0.04) and the dominant model (SMD=0.07, 95% CI=0.01-0.14, I2=22%, p=0.02) in the general population excluding the elderly. While abnormal folate concentrations are more common in elderly, no association between MTHFR A1298C polymorphism and peripheral blood folate concentration was found in the meta-analysis when elderly were included. CONCLUSIONS: This meta-analysis indicates that, in the general population excluding the elderly, the C allele of MTHFR 1298 polymorphism is associated with the risk for an increased folate concentration.


Asunto(s)
Ácido Fólico/sangre , Regulación Enzimológica de la Expresión Génica/fisiología , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Polimorfismo de Nucleótido Simple , Femenino , Ácido Fólico/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Estudios Observacionales como Asunto
20.
J Diabetes Investig ; 8(4): 560-570, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28002648

RESUMEN

AIMS/INTRODUCTION: Studies have been carried out to evaluate the correlation between TCF7L2 genetic polymorphisms and gestational diabetes mellitus (GDM) risk. However, the conclusions from these studies are incomplete, because partial single nucleotide polymorphisms (SNPs) were analyzed. We carried out a meta-analysis aimed to systematically evaluate TCF7L2 gene polymorphisms and GDM susceptibility in all population and racial/ethnic subgroups to afford a foundation for future research. MATERIALS AND METHODS: Published studies censoring TCF7L2 variants and GDM risk were captured from the EMBASE, PubMed, CNKI and Wanfang databases. The meta-analysis was processed using software of RevMan 5.2 and Stata13. The relationship between TCF7L2 polymorphism and GDM occurrence was evaluated by pooled odds ratios. Stratified analysis based on race/ethnicity was also carried out. The allele-specific odds ratios and 95% confidence intervals were counted, and based on homogeneity evaluated using the I2 -test, fixed- or random-effects pooled measures were selected. RESULTS: A total of 22 studies were covered, capturing eight TCF7L2 SNPs and involving 5,573 cases and 13,266 controls. Six of eight SNPs showed significant relationships with GDM occurrence, of which the SNPs rs7903146, rs12255372 and rs7901695 were the most powerful. Stratified analysis by race/ethnicity showed discrepant results in these three SNPs. In Caucasians and other races, all these SNPs were found to have a significant association with GDM risk, but in Asians, only SNP rs7903146 showed a significant association. CONCLUSIONS: Six of eight SNPs were found to have significant associations between TCF7L2 variants and GDM risk in the overall population, with the most powerful in SNPs being rs7903146, rs12255372 and rs7901695, but the contribution of these SNPs to GDM risk were variable among different racial/ethnic groups.


Asunto(s)
Diabetes Gestacional/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Diabetes Gestacional/etnología , Femenino , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Humanos , Polimorfismo de Nucleótido Simple , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA