Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.407
Filtrer
1.
Birth Defects Res ; 116(6): e2372, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38877667

RÉSUMÉ

OBJECTIVE: To determine the effect of maternal status in (plasma and red blood cell) folate, vitamin B12, homocysteine, and vitamin D, as well as their interaction with MTHFR (C677T and A1298C) and MTRR A66G polymorphisms, on maternal plasma docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) levels and the risk of neural tube defects (NTDs). METHODS: ARA, EPA, and DHA composition was assessed using capillary gas chromatography. RESULTS: ARA and DHA levels were higher in controls than in case mothers for low plasma folate status. For low red blood cell folate status, DHA levels were higher in controls than in case mothers. For high homocysteine levels, ARA and DHA levels were higher in controls than in case mothers. NTD mothers had lower EPA and DHA levels for low vitamin B12 levels. NTD mothers had lower DHA levels for low vitamin D levels. For low plasma folate status, DHA levels in the MTHFR C677T gene and ARA and EPA levels in MTHFR A1298C gene were different among the three genotypes in case mothers. DHA levels in the MTHFR C677T gene were different among the three genotypes in case mothers for both low and high homocysteine levels. For low vitamin B12 levels, ARA and DHA levels were different among the three genotypes of the MTHFR C677T gene in case mothers. In the MTHFR C677T gene, ARA and DHA levels were different among the three genotypes in case mothers for low vitamin D levels. CONCLUSIONS: More advanced research is required to verify a suitable biochemical parameter status in relation to the genotypes in pregnant women.


Sujet(s)
Acide arachidonique , Acide docosahexaénoïque , Acide eicosapentanoïque , Acide folique , Methylenetetrahydrofolate reductase (NADPH2) , Anomalies du tube neural , Humains , Acide eicosapentanoïque/sang , Acide docosahexaénoïque/sang , Femelle , Anomalies du tube neural/génétique , Acide arachidonique/sang , Acide arachidonique/métabolisme , Acide folique/sang , Adulte , Tunisie , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Homocystéine/sang , Homocystéine/génétique , Grossesse , Vitamine B12/sang , Études cas-témoins , Génotype , Vitamine D/sang , Vitamine D/génétique
2.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891776

RÉSUMÉ

Neural tube defects (NTDs), which are caused by impaired embryonic neural tube closure, are one of the most serious and common birth defects. Peptidyl-prolyl cis/trans isomerase 1 (Pin1) is a prolyl isomerase that uniquely regulates cell signaling by manipulating protein conformation following phosphorylation, although its involvement in neuronal development remains unknown. In this study, we explored the involvement of Pin1 in NTDs and its potential mechanisms both in vitro and in vivo. The levels of Pin1 expression were reduced in NTD models induced by all-trans retinoic acid (Atra). Pin1 plays a significant role in regulating the apoptosis, proliferation, differentiation, and migration of neurons. Moreover, Pin1 knockdown significantly was found to exacerbate oxidative stress (OS) and endoplasmic reticulum stress (ERs) in neuronal cells. Further studies showed that the Notch1-Nrf2 signaling pathway may participate in Pin1 regulation of NTDs, as evidenced by the inhibition and overexpression of the Notch1-Nrf2 pathway. In addition, immunofluorescence (IF), co-immunoprecipitation (Co-IP), and GST pull-down experiments also showed that Pin1 interacts directly with Notch1 and Nrf2. Thus, our study suggested that the knocking down of Pin1 promotes NTD progression by inhibiting the activation of the Notch1-Nrf2 signaling pathway, and it is possible that this effect is achieved by disrupting the interaction of Pin1 with Notch1 and Nrf2, affecting their proteostasis. Our research identified that the regulation of Pin1 by retinoic acid (RA) and its involvement in the development of NTDs through the Notch1-Nrf2 axis could enhance our comprehension of the mechanism behind RA-induced brain abnormalities.


Sujet(s)
NIMA-interacting peptidylprolyl isomerase , Anomalies du tube neural , Récepteur Notch1 , Trétinoïne , Trétinoïne/métabolisme , Trétinoïne/pharmacologie , NIMA-interacting peptidylprolyl isomerase/métabolisme , NIMA-interacting peptidylprolyl isomerase/génétique , Animaux , Souris , Anomalies du tube neural/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/induit chimiquement , Récepteur Notch1/métabolisme , Récepteur Notch1/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Femelle , Tube neural/métabolisme , Tube neural/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Humains
3.
Genesis ; 62(3): e23602, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38721990

RÉSUMÉ

Cilia play a key role in the regulation of signaling pathways required for embryonic development, including the proper formation of the neural tube, the precursor to the brain and spinal cord. Forward genetic screens were used to generate mouse lines that display neural tube defects (NTD) and secondary phenotypes useful in interrogating function. We describe here the L3P mutant line that displays phenotypes of disrupted Sonic hedgehog signaling and affects the initiation of cilia formation. A point mutation was mapped in the L3P line to the gene Rsg1, which encodes a GTPase-like protein. The mutation lies within the GTP-binding pocket and disrupts the highly conserved G1 domain. The mutant protein and other centrosomal and IFT proteins still localize appropriately to the basal body of cilia, suggesting that RSG1 GTPase activity is not required for basal body maturation but is needed for a downstream step in axonemal elongation.


Sujet(s)
Cils vibratiles , Tube neural , Animaux , Souris , Cils vibratiles/métabolisme , Cils vibratiles/génétique , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Tube neural/embryologie , Tube neural/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Mutation ponctuelle , Transduction du signal
4.
Birth Defects Res ; 116(5): e2333, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38716581

RÉSUMÉ

OBJECTIVE: This study aims to determine if 5,10-methylenetetrahydrofolate reductase (MTHFR C677T and A1298C) and methionine synthase reductase (MTRR A66G) gene polymorphisms were associated with fatty acid (FA) levels in mothers of fetuses with neural tube defects (NTDs) and whether these associations were modified by environmental factors. METHODS: Plasma FA composition was assessed using capillary gas chromatography. Concentrations of studied FA were compared between 42 mothers of NTDs fetuses and 30 controls as a function of each polymorphism by the Kruskal-Wallis nonparametric test. RESULTS: In MTHFR gene C677T polymorphism, cases with (CT + TT) genotype had lower monounsaturated FAs (MUFA) and omega-3 polyunsaturated FA (n-3 PUFA) levels, but higher omega-6 polyunsaturated FAs (n-6 PUFA) and omega-6 polyunsaturated FAs: omega-3 polyunsaturated FAs (n-6:n-3) ratio levels. In MTRR gene A66G polymorphism, cases with (AG + GG) genotype had lower MUFA levels, but higher PUFA and n-6 PUFA levels. Controls with (AG + GG) genotype had lower n-6 PUFA levels. In MTHFR gene C677T polymorphism, cases with smoking spouses and (CT + TT) genotype had lower MUFA and n-3 PUFA levels, but higher PUFA, n-6 PUFA, and n-6:n-3 ratio levels. Cases with (CT + TT) genotype and who used sauna during pregnancy had lower n-3 PUFA levels. In MTRR gene A66G polymorphism, cases with (AG + GG) genotype and who used sauna during pregnancy had higher PUFA and n-6 PUFA levels. CONCLUSIONS: Further research is required to clarify the association of FA metabolism and (MTHFR, MTRR) polymorphisms with NTDs.


Sujet(s)
Acides gras , Ferredoxine-NADP reductase , Prédisposition génétique à une maladie , Methylenetetrahydrofolate reductase (NADPH2) , Anomalies du tube neural , Polymorphisme de nucléotide simple , Humains , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Methylenetetrahydrofolate reductase (NADPH2)/métabolisme , Femelle , Anomalies du tube neural/génétique , Ferredoxine-NADP reductase/génétique , Ferredoxine-NADP reductase/métabolisme , Adulte , Acides gras/métabolisme , Polymorphisme de nucléotide simple/génétique , Grossesse , Génotype , Études cas-témoins , Facteurs de risque , Acides gras omega-3/métabolisme , Acides gras omega-3/génétique , Acides gras omega-6/métabolisme , Acides gras omega-6/sang , Études d'associations génétiques/méthodes
5.
Cell Biol Toxicol ; 40(1): 35, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38771546

RÉSUMÉ

Neural tube defects (NTDs) represent a prevalent and severe category of congenital anomalies in humans. Cadmium (Cd) is an environmental teratogen known to cause fetal NTDs. However, its underlying mechanisms remain elusive. This study aims to investigate the therapeutic potential of lipophagy in the treatment of NTDs, providing valuable insights for future strategies targeting lipophagy activation as a means to mitigate NTDs.We successfully modeled NTDs by Cd exposure during pregnancy. RNA sequencing was employed to investigate the transcriptomic alterations and functional enrichment of differentially expressed genes in NTD placental tissues. Subsequently, pharmacological/genetic (Atg5-/- placentas) experiments confirmed that inducing placental lipophagy can alleviate Cd induced-NTDs. We found that Cd exposure caused NTDs. Further analyzed transcriptomic data from the placentas with NTDs which revealed significant downregulation of low-density lipoprotein receptor associated protein 1(Lrp1) gene expression responsible for positive regulation of low-density lipoprotein cholesterol (LDL-C) transport. Correspondingly, there was an increase in maternal serum/placenta/amniotic fluid LDL-C content. Subsequently, we have discovered that Cd exposure activated placental lipophagy. Pharmacological/genetic (Atg5-/- placentas) experiments confirmed that inducing placental lipophagy can alleviate Cd induced-NTDs. Furthermore, our findings demonstrate that activation of placental lipophagy effectively counteracts the Cd-induced elevation in LDL-C levels. Lipophagy serves to mitigate Cd-induced NTDs by reducing LDL-C levels within mouse placentas.


Sujet(s)
Cadmium , Cholestérol LDL , Anomalies du tube neural , Placenta , Femelle , Animaux , Grossesse , Placenta/métabolisme , Placenta/effets des médicaments et des substances chimiques , Anomalies du tube neural/génétique , Anomalies du tube neural/induit chimiquement , Anomalies du tube neural/métabolisme , Souris , Cadmium/toxicité , Cholestérol LDL/sang , Protéine-1 apparentée au récepteur des LDL/génétique , Protéine-1 apparentée au récepteur des LDL/métabolisme , Souris de lignée C57BL , Souris knockout
6.
Development ; 151(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38682273

RÉSUMÉ

Neurulation is a highly synchronized biomechanical process leading to the formation of the brain and spinal cord, and its failure leads to neural tube defects (NTDs). Although we are rapidly learning the genetic mechanisms underlying NTDs, the biomechanical aspects are largely unknown. To understand the correlation between NTDs and tissue stiffness during neural tube closure (NTC), we imaged an NTD murine model using optical coherence tomography (OCT), Brillouin microscopy and confocal fluorescence microscopy. Here, we associate structural information from OCT with local stiffness from the Brillouin signal of embryos undergoing neurulation. The stiffness of neuroepithelial tissues in Mthfd1l null embryos was significantly lower than that of wild-type embryos. Additionally, exogenous formate supplementation improved tissue stiffness and gross embryonic morphology in nullizygous and heterozygous embryos. Our results demonstrate the significance of proper tissue stiffness in normal NTC and pave the way for future studies on the mechanobiology of normal and abnormal embryonic development.


Sujet(s)
Anomalies du tube neural , Tube neural , Neurulation , Tomographie par cohérence optique , Animaux , Tomographie par cohérence optique/méthodes , Souris , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Anomalies du tube neural/anatomopathologie , Tube neural/métabolisme , Neurulation/génétique , Methylenetetrahydrofolate Dehydrogenase (NADP)/génétique , Methylenetetrahydrofolate Dehydrogenase (NADP)/métabolisme , Formiates/métabolisme , Embryon de mammifère/métabolisme , Femelle , Formate-tetrahydrofolate ligase/génétique , Formate-tetrahydrofolate ligase/métabolisme , Mutation/génétique , Phénomènes biomécaniques , Microscopie confocale , Souris knockout
7.
Reprod Toxicol ; 125: 108576, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38479591

RÉSUMÉ

Folate is a micronutrient essential for DNA synthesis, cell division, fetal growth and development. Folate deficiency leads to genomic instability. Inadequate intake of folate during conception may lead to neural tube defects (NTDs) in the offspring. Folate influences the DNA methylation, histone methylation and homocysteine mediated gene methylation. DNA methylation influences the expression of microRNAs (miRNAs). Folate deficiency may be associated with miRNAs misregulation leading to NTDs. Mitochondrial epigenetics and folate metabolism has proved to be involved in embryogenesis and neural tube development. Folate related genetic variants also cause the occurrence of NTDs. Unmetabolized excessive folate may affect health adversely. Hence estimation of folate levels in the blood plays an important role in high-risk cases.


Sujet(s)
Carence en acide folique , microARN , Anomalies du tube neural , Humains , Acide folique , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Carence en acide folique/complications , Carence en acide folique/génétique , Carence en acide folique/métabolisme , Épigenèse génétique , Méthylation de l'ADN , microARN/génétique , Tube neural/métabolisme
8.
Hum Genet ; 143(3): 263-277, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38451291

RÉSUMÉ

Neural tube defects (NTDs) are severe malformations of the central nervous system that arise from failure of neural tube closure. HECTD1 is an E3 ubiquitin ligase required for cranial neural tube closure in mouse models. NTDs in the Hectd1 mutant mouse model are due to the failure of cranial mesenchyme morphogenesis during neural fold elevation. Our earlier research has linked increased extracellular heat shock protein 90 (eHSP90) secretion to aberrant cranial mesenchyme morphogenesis in the Hectd1 model. Furthermore, overexpression of HECTD1 suppresses stress-induced eHSP90 secretion in cell lines. In this study, we report the identification of five rare HECTD1 missense sequence variants in NTD cases. The variants were found through targeted next-generation sequencing in a Chinese cohort of 352 NTD cases and 224 ethnically matched controls. We present data showing that HECTD1 is a highly conserved gene, extremely intolerant to loss-of-function mutations and missense changes. To evaluate the functional consequences of NTD-associated missense variants, functional assays in HEK293T cells were performed to examine protein expression and the ability of HECTD1 sequence variants to suppress eHSP90 secretion. One NTD-associated variant (A1084T) had significantly reduced expression in HEK293T cells. All five NTD-associated variants (p.M392V, p.T801I, p.I906V, p.A1084T, and p.P1835L) reduced regulation of eHSP90 secretion by HECTD1, while a putative benign variant (p.P2474L) did not. These findings are the first association of HECTD1 sequence variation with NTDs in humans.


Sujet(s)
Mutation faux-sens , Anomalies du tube neural , Ubiquitin-protein ligases , Humains , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Anomalies du tube neural/génétique , Cellules HEK293 , Protéines du choc thermique HSP90/génétique , Protéines du choc thermique HSP90/métabolisme , Femelle , Mâle , Souris , Animaux
9.
J Transl Med ; 22(1): 257, 2024 Mar 09.
Article de Anglais | MEDLINE | ID: mdl-38461288

RÉSUMÉ

BACKGROUND: Neural Tube Defects (NTDs) are congenital malformations of the central nervous system resulting from the incomplete closure of the neural tube during early embryonic development. Neuroinflammation refers to the inflammatory response in the nervous system, typically resulting from damage to neural tissue. Immune-related processes have been identified in NTDs, however, the detailed relationship and underlying mechanisms between neuroinflammation and NTDs remain largely unclear. In this study, we utilized integrated multi-omics analysis to explore the role of neuroinflammation in NTDs and identify potential prenatal diagnostic markers using a murine model. METHODS: Nine public datasets from Gene Expression Omnibus (GEO) and ArrayExpress were mined using integrated multi-omics analysis to characterize the molecular landscape associated with neuroinflammation in NTDs. Special attention was given to the involvement of macrophages in neuroinflammation within amniotic fluid, as well as the dynamics of macrophage polarization and their interactions with neural cells at single-cell resolution. We also used qPCR assay to validate the key TFs and candidate prenatal diagnostic genes identified through the integrated analysis in a retinoic acid-induced NTDs mouse model. RESULTS: Our analysis indicated that neuroinflammation is a critical pathological feature of NTDs, regulated both transcriptionally and epigenetically within central nervous system tissues. Key alterations in gene expression and pathways highlighted the crucial role of STATs molecules in the JAK-STAT signaling pathway in regulating NTDs-associated neuroinflammation. Furthermore, single-cell resolution analysis revealed significant polarization of macrophages and their interaction with neural cells in amniotic fluid, underscoring their central role in mediating neuroinflammation associated with NTDs. Finally, we identified a set of six potential prenatal diagnostic genes, including FABP7, CRMP1, SCG3, SLC16A10, RNASE6 and RNASE1, which were subsequently validated in a murine NTDs model, indicating their promise as prospective markers for prenatal diagnosis of NTDs. CONCLUSIONS: Our study emphasizes the pivotal role of neuroinflammation in the progression of NTDs and underlines the potential of specific inflammatory and neural markers as novel prenatal diagnostic tools. These findings provide important clues for further understanding the underlying mechanisms between neuroinflammation and NTDs, and offer valuable insights for the future development of prenatal diagnostics.


Sujet(s)
Multi-omique , Anomalies du tube neural , Grossesse , Femelle , Animaux , Souris , Maladies neuro-inflammatoires , Études prospectives , Anomalies du tube neural/diagnostic , Anomalies du tube neural/génétique , Anomalies du tube neural/induit chimiquement , Système nerveux central/anatomopathologie
10.
Nature ; 626(8001): 1042-1048, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38418917

RÉSUMÉ

The loss of the tail is among the most notable anatomical changes to have occurred along the evolutionary lineage leading to humans and to the 'anthropomorphous apes'1-3, with a proposed role in contributing to human bipedalism4-6. Yet, the genetic mechanism that facilitated tail-loss evolution in hominoids remains unknown. Here we present evidence that an individual insertion of an Alu element in the genome of the hominoid ancestor may have contributed to tail-loss evolution. We demonstrate that this Alu element-inserted into an intron of the TBXT gene7-9-pairs with a neighbouring ancestral Alu element encoded in the reverse genomic orientation and leads to a hominoid-specific alternative splicing event. To study the effect of this splicing event, we generated multiple mouse models that express both full-length and exon-skipped isoforms of Tbxt, mimicking the expression pattern of its hominoid orthologue TBXT. Mice expressing both Tbxt isoforms exhibit a complete absence of the tail or a shortened tail depending on the relative abundance of Tbxt isoforms expressed at the embryonic tail bud. These results support the notion that the exon-skipped transcript is sufficient to induce a tail-loss phenotype. Moreover, mice expressing the exon-skipped Tbxt isoform develop neural tube defects, a condition that affects approximately 1 in 1,000 neonates in humans10. Thus, tail-loss evolution may have been associated with an adaptive cost of the potential for neural tube defects, which continue to affect human health today.


Sujet(s)
Épissage alternatif , Évolution moléculaire , Hominidae , Protéines à domaine boîte-T , Queue , Animaux , Humains , Souris , Épissage alternatif/génétique , Séquences Alu/génétique , Modèles animaux de maladie humaine , Génome/génétique , Hominidae/anatomie et histologie , Hominidae/génétique , Introns/génétique , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Phénotype , Isoformes de protéines/déficit , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéines à domaine boîte-T/déficit , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Queue/anatomie et histologie , Queue/embryologie , Exons/génétique
11.
Nat Commun ; 15(1): 1642, 2024 Feb 22.
Article de Anglais | MEDLINE | ID: mdl-38388461

RÉSUMÉ

Folate supplementation reduces the occurrence of neural tube defects (NTDs), birth defects consisting in the failure of the neural tube to form and close. The mechanisms underlying NTDs and their prevention by folate remain unclear. Here we show that folate receptor 1 (FOLR1) is necessary for the formation of neural tube-like structures in human-cell derived neural organoids. FOLR1 knockdown in neural organoids and in Xenopus laevis embryos leads to NTDs that are rescued by pteroate, a folate precursor that is unable to participate in metabolism. We demonstrate that FOLR1 interacts with and opposes the function of CD2-associated protein, molecule essential for apical endocytosis and turnover of C-cadherin in neural plate cells. In addition, folates increase Ca2+ transient frequency, suggesting that folate and FOLR1 signal intracellularly to regulate neural plate folding. This study identifies a mechanism of action of folate distinct from its vitamin function during neural tube formation.


Sujet(s)
Acide folique , Anomalies du tube neural , Humains , Acide folique/métabolisme , Tube neural/métabolisme , Récepteur-1 des folates/génétique , Récepteur-1 des folates/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Plaque neurale/métabolisme
12.
Epigenomics ; 16(6): 419-426, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38410929

RÉSUMÉ

Neural tube defects (NTDs) are the most common congenital anomalies of the CNS. It is widely appreciated that both genetic and environmental factors contribute to their etiology. The inability to ascribe clear genetic patterns of inheritance to various NTD phenotypes suggests it is possible that epigenetic mechanisms are involved in the etiology of NTDs. In this context, the contribution of DNA methylation as an underlying contributing factor to the etiology of NTDs has been extensively reviewed. Here, an updated accounting of the evidence linking post-translational histone modifications to these birth defects, relying heavily upon studies in humans, and the possible molecular implications inferred from reports based on cellular and animal models, are presented.


Sujet(s)
Histone , Anomalies du tube neural , Animaux , Humains , Histone/métabolisme , Code histone , Anomalies du tube neural/génétique , Épigenèse génétique , Méthylation de l'ADN
13.
Genome Biol ; 25(1): 19, 2024 01 15.
Article de Anglais | MEDLINE | ID: mdl-38225631

RÉSUMÉ

BACKGROUND: Neural tube defects (NTDs) are caused by genetic and environmental factors. ARMC5 is part of a novel ubiquitin ligase specific for POLR2A, the largest subunit of RNA polymerase II (Pol II). RESULTS: We find that ARMC5 knockout mice have increased incidence of NTDs, such as spina bifida and exencephaly. Surprisingly, the absence of ARMC5 causes the accumulation of not only POLR2A but also most of the other 11 Pol II subunits, indicating that the degradation of the whole Pol II complex is compromised. The enlarged Pol II pool does not lead to generalized Pol II stalling or a generalized decrease in mRNA transcription. In neural progenitor cells, ARMC5 knockout only dysregulates 106 genes, some of which are known to be involved in neural tube development. FOLH1, critical in folate uptake and hence neural tube development, is downregulated in the knockout intestine. We also identify nine deleterious mutations in the ARMC5 gene in 511 patients with myelomeningocele, a severe form of spina bifida. These mutations impair the interaction between ARMC5 and Pol II and reduce Pol II ubiquitination. CONCLUSIONS: Mutations in ARMC5 increase the risk of NTDs in mice and humans. ARMC5 is part of an E3 controlling the degradation of all 12 subunits of Pol II under physiological conditions. The Pol II pool size might have effects on NTD pathogenesis, and some of the effects might be via the downregulation of FOLH1. Additional mechanistic work is needed to establish the causal effect of the findings on NTD pathogenesis.


Sujet(s)
Protéines à domaine armadillo , Anomalies du tube neural , Dysraphie spinale , Animaux , Humains , Souris , Protéines à domaine armadillo/génétique , Acide folique/métabolisme , Souris knockout , Mutation , Anomalies du tube neural/génétique , Anomalies du tube neural/épidémiologie , Dysraphie spinale/génétique
14.
Zool Res ; 45(2): 233-241, 2024 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-38287904

RÉSUMÉ

Neural tube defects (NTDs) are severe congenital neurodevelopmental disorders arising from incomplete neural tube closure. Although folate supplementation has been shown to mitigate the incidence of NTDs, some cases, often attributable to genetic factors, remain unpreventable. The SHROOM3 gene has been implicated in NTD cases that are unresponsive to folate supplementation; at present, however, the underlying mechanism remains unclear. Neural tube morphogenesis is a complex process involving the folding of the planar epithelium of the neural plate. To determine the role of SHROOM3 in early developmental morphogenesis, we established a neuroepithelial organoid culture system derived from cynomolgus monkeys to closely mimic the in vivo neural plate phase. Loss of SHROOM3 resulted in shorter neuroepithelial cells and smaller nuclei. These morphological changes were attributed to the insufficient recruitment of cytoskeletal proteins, namely fibrous actin (F-actin), myosin II, and phospho-myosin light chain (PMLC), to the apical side of the neuroepithelial cells. Notably, these defects were not rescued by folate supplementation. RNA sequencing revealed that differentially expressed genes were enriched in biological processes associated with cellular and organ morphogenesis. In summary, we established an authentic in vitro system to study NTDs and identified a novel mechanism for NTDs that are unresponsive to folate supplementation.


Sujet(s)
Protéines du cytosquelette , Anomalies du tube neural , Animaux , Protéines du cytosquelette/métabolisme , Tube neural/métabolisme , Macaca fascicularis , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Anomalies du tube neural/médecine vétérinaire , Cellules neuroépithéliales/métabolisme , Acide folique/métabolisme , Organoïdes , Cytosquelette
15.
Am J Obstet Gynecol ; 230(2): 254.e1-254.e13, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37531989

RÉSUMÉ

BACKGROUND: Hyperglycemia from pregestational diabetes mellitus induces neural tube defects in the developing fetus. Folate supplementation is the only effective way to prevent neural tube defects; however, some cases of neural tube defects are resistant to folate. Excess folate has been linked to higher maternal cancer risk and infant allergy. Therefore, additional interventions are needed. Understanding the mechanisms underlying maternal diabetes mellitus-induced neural tube defects can identify potential targets for preventing such defects. Despite not yet being in clinical use, growing evidence suggests that microRNAs are important intermediates in embryonic development and can serve as both biomarkers and drug targets for disease intervention. Our previous studies showed that maternal diabetes mellitus in vivo activates the inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in the developing embryo and that a high glucose condition in vitro reduces microRNA-322 (miR-322) levels. IRE1α is an RNA endonuclease; however, it is unknown whether IRE1α targets and degrades miR-322 specifically or whether miR-322 degradation leads to neural tube defects via apoptosis. We hypothesize that IRE1α can inhibit miR-322 in maternal diabetes mellitus-induced neural tube defects and that restoring miR-322 expression in developing neuroepithelium ameliorates neural tube defects. OBJECTIVE: This study aimed to identify potential targets for preventing maternal diabetes mellitus-induced neural tube defects and to investigate the roles and relationship of a microRNA and an RNA endonuclease in mouse embryos exposed to maternal diabetes mellitus. STUDY DESIGN: To determine whether miR-322 reduction is necessary for neural tube defect formation in pregnancies complicated by diabetes mellitus, male mice carrying a transgene expressing miR-322 were mated with nondiabetic or diabetic wide-type female mice to generate embryos with or without miR-322 overexpression. At embryonic day 8.5 when the neural tube is not yet closed, embryos were harvested for the assessment of 3 miR-322 transcripts (primary, precursor, and mature miR-322), tumor necrosis factor receptor-associated factor 3 (TRAF3), and neuroepithelium cell survival. Neural tube defect incidences were determined in embryonic day 10.5 embryos when the neural tube should be closed if there is no neural tube defect formation. To identify which miR-322 transcript is affected by maternal diabetes mellitus and high glucose conditions, 3 miR-322 transcripts were assessed in embryos from dams with or without diabetes mellitus and in C17.2 mouse neural stem cells treated with different concentrations of glucose and at different time points. To determine whether the endonuclease IRE1α targets miR-322, small interfering RNA knockdown of IRE1α or overexpression of inositol-requiring transmembrane kinase/endoribonuclease 1α by DNA plasmid transfection was used to determine the effect of IRE1α deficiency or overexpression on miR-322 expression. RNA immunoprecipitation was performed to reveal the direct targets of inositol-requiring transmembrane kinase/endoribonuclease 1α. RESULTS: Maternal diabetes mellitus suppressed miR-322 expression in the developing neuroepithelium. Restoring miR-322 expression in the neuroepithelium blocked maternal diabetes mellitus-induced caspase-3 and caspase-8 cleavage and cell apoptosis, leading to a neural tube defect reduction. Reversal of maternal diabetes mellitus-inhibited miR-322 via transgenic overexpression prevented TRAF3 up-regulation in embryos exposed to maternal diabetes mellitus. Activated IRE1α acted as an endonuclease and degraded precursor miR-322, resulting in mature miR-322 reduction. CONCLUSION: This study supports the crucial role of the IRE1α-microRNA-TRAF3 circuit in the induction of neuroepithelial cell apoptosis and neural tube defect formation in pregnancies complicated by diabetes mellitus and identifies IRE1α and miR-322 as potential targets for preventing maternal diabetes mellitus-induced neural tube defects.


Sujet(s)
Diabète expérimental , Diabète gestationnel , microARN , Anomalies du tube neural , Grossesse chez les diabétiques , Humains , Grossesse , Mâle , Femelle , Souris , Animaux , microARN/génétique , microARN/métabolisme , Facteur-3 associé aux récepteurs de TNF/métabolisme , Endoribonucleases/génétique , Endoribonucleases/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Diabète expérimental/génétique , Diabète expérimental/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/anatomopathologie , Grossesse chez les diabétiques/génétique , Grossesse chez les diabétiques/métabolisme , Diabète gestationnel/génétique , Glucose , Acide folique , Inositol
16.
FASEB J ; 38(1): e23346, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38095297

RÉSUMÉ

Folate deficiency contribute to neural tube defects (NTDs) which could be rescued by folate supplementation. However, the underlying mechanisms are still not fully understood. Besides, there is considerable controversy concerning the forms of folate used for supplementation. To address this controversy, we prepared culture medium with different forms of folate, folic acid (FA), and 5-methyltetrahydrofolate (5mTHF), at concentrations of 5 µM, 500 nM, 50 nM, and folate free, respectively. Mouse embryonic fibroblasts (MEFs) were treated with different folates continuously for three passages, and cell proliferation and F-actin were monitored. We determined that compared to 5mTHF, FA showed stronger effects on promoting cell proliferation and F-actin formation. We also found that FOLR1 protein level was positively regulated by folate concentration and the non-canonical Wnt/planar cell polarity (PCP) pathway signaling was significantly enriched among different folate conditions in RNA-sequencing analyses. We demonstrated for the first time that FOLR1 could promote the transcription of Vangl2, one of PCP core genes. The transcription of Vangl2 was down-regulated under folate-deficient condition, which resulted in a decrease in PCP activity and F-actin formation. In summary, we identified a distinct advantage of FA in cell proliferation and F-actin formation over 5mTHF, as well as demonstrating that FOLR1 could promote transcription of Vangl2 and provide a new mechanism by which folate deficiency can contribute to the etiology of NTDs.


Sujet(s)
Carence en acide folique , Anomalies du tube neural , Animaux , Souris , Acide folique/métabolisme , Actines/métabolisme , Récepteur-1 des folates/génétique , Récepteur-1 des folates/métabolisme , Polarité de la cellule/génétique , Fibroblastes/métabolisme , Voie de signalisation Wnt , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Carence en acide folique/métabolisme
17.
Epigenetics Chromatin ; 16(1): 50, 2023 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-38093377

RÉSUMÉ

BACKGROUND: Neural tube defects (NTDs) are one of the most severe congenital abnormalities characterized by failures of the neural tube to close during early embryogenesis. Maternal folate deficiency could impact the occurrence of NTDs, however, the mechanisms involved in the cause of NTDs are poorly defined. RESULTS: Here, we report that histone H3 methyltransferase disruptor of telomeric silencing 1-like (DOT1L) expression was significantly downregulated, and low levels of H3K79me2 were found in the corresponding NTDs samples with their maternal serum folate under low levels. Using ChIP-seq assays, we found that a decrease of H3K79me2 downregulates the expression of Shh and Sufu in mouse embryonic stem cells (mESC) under folate deficiency. Interestingly, folate antagonist methotrexate treatment led to attenuation of H3K79me2 due to Dot1l, affecting Shh and Sufu genes regulation. Upon further analysis, we find that the genes Shh and Sufu are both downregulated in the brain tissues of mice and humans with NTDs. There was a positive correlation between the transcription levels of Shh, Sufu and the protein levels of DOT1L by Pearson correlation analysis. CONCLUSION: Our results indicate that abnormal Shh and Sufu genes expression reduced by aberrant Dot1l-mediated H3K79me2 levels could be the cause of NTDs occurrence.


Sujet(s)
Histone , Anomalies du tube neural , Souris , Humains , Animaux , Histone/métabolisme , Anomalies du tube neural/génétique , Anomalies du tube neural/métabolisme , Méthylation , Acide folique/métabolisme , Expression des gènes , Protéines Hedgehog/génétique , Protéines Hedgehog/métabolisme , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/métabolisme
18.
Biomed Environ Sci ; 36(9): 837-849, 2023 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-37803896

RÉSUMÉ

Objective: Circular RNAs (circRNAs) participate in several important pathological processes and have been used in the diagnosis and treatment of various diseases. This study aimed to investigate the role of circRNAs in neural tube defects (NTDs). Method: We characterized circRNA-associated competitive endogenous RNA (ceRNA) networks in brain tissue of low folate -induced NTDs mouse at embryonic day 13.5 by high-throughput sequencing. The expression levels of Circzfp644, miR-20-5p and Gas7 were detected by RT-PCR. Gas7 and Circzfp644 functions were determined by miRNA-mimics and inhibitors in mouse teratocarcinoma cells (F9 cells), and luciferase gene reporter assay was assessed in the F9 cells. In addition, the expression levels of Circzfp644, miR-20-5p and Gas7 were determined by Nanostring in human NTDs tissues. Results: We detected 57 circRNA transcripts, 16 miRNAs, and 148 mRNAs that were significantly dysregulated in NTDs brain tissues compared with their expression levels in control (normal) tissues. Circzfp644 shared miRNA response elements with the growth arrest specific 7 ( Gas7) gene and competitively bound with miR-20-5p to increase the expression of Gas7. Downregulation of Circzfp644 and Gas7 and upregulation of miR-20-5p were found in human NTD tissue. Conclusion: This study provides new perspectives on the role of circRNAs in nervous system development and the pathogenesis of NTDs.


Sujet(s)
microARN , Anomalies du tube neural , Humains , Animaux , Souris , ARN circulaire/génétique , microARN/génétique , microARN/métabolisme , Régulation négative , Anomalies du tube neural/génétique , Acide folique
19.
J Med Genet ; 60(12): 1146-1152, 2023 Nov 27.
Article de Anglais | MEDLINE | ID: mdl-37775263

RÉSUMÉ

Congenital vertebral malformations (CVMs) and neural tube defects (NTDs) are common birth defects affecting the spine and nervous system, respectively, due to defects in somitogenesis and neurulation. Somitogenesis and neurulation rely on factors secreted from neighbouring tissues and the integrity of the axial structure. Crucial signalling pathways like Wnt, Notch and planar cell polarity regulate somitogenesis and neurulation with significant crosstalk. While previous studies suggest an association between CVMs and NTDs, the exact mechanism underlying this relationship remains unclear. In this review, we explore embryonic development, signalling pathways and clinical phenotypes involved in the association between CVMs and NTDs. Moreover, we provide a summary of syndromes that exhibit occurrences of both CVMs and NTDs. We aim to provide insights into the potential mechanisms underlying the association between CVMs and NTDs, thereby facilitating clinical diagnosis and management of these anomalies.


Sujet(s)
Anomalies du tube neural , Femelle , Grossesse , Humains , Anomalies du tube neural/épidémiologie , Anomalies du tube neural/génétique , Rachis/métabolisme , Développement embryonnaire , Neurulation/génétique , Transduction du signal/génétique
20.
Birth Defects Res ; 115(18): 1770-1779, 2023 11 01.
Article de Anglais | MEDLINE | ID: mdl-37776548

RÉSUMÉ

BACKGROUND: Maternal diabetes increases the risk for neural tube defects (NTDs). It is unclear if miRNAs, senescence, and DNA damage are involved in this process. In this study, we used neural stem cells as an in vitro proxy of embryonic neuroepithelium to investigate whether high glucose triggers neural stem cell senescence and DNA damage by upregulating miR-200c, which may be responsible for NTDs. METHODS: C17.2 neural stem cells were cultured with normal glucose (5 mM) or high glucose (≥16.7 mM) at different doses and time points for detecting miR-200c levels, markers of senescence and DNA damage. Neural stem cells were exposed to antioxidant SOD1 mimetic Tempol and high glucose for 48 h to test roles of oxidative stress on the miR-200c, senescence, and DNA damage levels. An miR-200c mimic and an inhibitor were transfected into neural stem cells to increase or decrease miR-200c activities. RESULTS: High glucose upregulated miR-200c in neural stem cells. A time course study of the effect of high glucose revealed that miR-200c initially increased at 12 h and reached its zenith at 18 h. Tempol reduced miR-200c levels caused by high glucose. High glucose induced markers of senescence and DNA damage in neural stem cells. Tempol abolished high glucose-induced markers of senescence and DNA damage. The miR-200c inhibitor suppressed high glucose-induced markers of senescence and DNA damage. Treatment with miR-200c mimic imitates high glucose-induced markers of senescence and DNA damage. CONCLUSIONS: We show that high glucose increases miR-200c, which contributes to cellular senescence and DNA damage in neural stem cells and provides a potential pathway for maternal diabetes-induced neural tube defects.


Sujet(s)
Diabète gestationnel , microARN , Cellules souches neurales , Anomalies du tube neural , Grossesse , Femelle , Humains , Cellules souches neurales/métabolisme , Vieillissement de la cellule/génétique , microARN/génétique , Anomalies du tube neural/génétique , Glucose/pharmacologie , Glucose/métabolisme , Altération de l'ADN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...