Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Commun ; 15(1): 1642, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38388461

RESUMO

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.


Assuntos
Ácido Fólico , Defeitos do Tubo Neural , Humanos , Ácido Fólico/metabolismo , Tubo Neural/metabolismo , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Placa Neural/metabolismo
2.
Zool Res ; 45(2): 233-241, 2024 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-38287904

RESUMO

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.


Assuntos
Proteínas do Citoesqueleto , Defeitos do Tubo Neural , Animais , Proteínas do Citoesqueleto/metabolismo , Tubo Neural/metabolismo , Macaca fascicularis , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/veterinária , Células Neuroepiteliais/metabolismo , Ácido Fólico/metabolismo , Organoides , Citoesqueleto
4.
Sci Rep ; 7(1): 5182, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701710

RESUMO

SR-BI is the main receptor for high density lipoproteins (HDL) and mediates the bidirectional transport of lipids, such as cholesterol and vitamin E, between these particles and cells. During early development, SR-BI is expressed in extraembryonic tissue, specifically in trophoblast giant cells in the parietal yolk sac. We previously showed that approximately 50% of SR-BI-/- embryos fail to close the anterior neural tube and develop exencephaly, a perinatal lethal condition. Here, we evaluated the role of SR-BI in embryonic vitamin E uptake during murine neural tube closure. Our results showed that SR-BI-/- embryos had a very low vitamin E content in comparison to SR-BI+/+ embryos. Whereas SR-BI-/- embryos with closed neural tubes (nSR-BI-/-) had high levels of reactive oxygen species (ROS), intermediate ROS levels between SR-BI+/+ and nSR-BI-/- embryos were detected in SR-BI-/- with NTD (NTD SR-BI-/-). Reduced expression of Pax3, Alx1 and Alx3 genes was found in NTD SR-BI-/- embryos. Maternal α-tocopherol dietary supplementation prevented NTD almost completely (from 54% to 2%, p < 0.001) in SR-BI-/- embryos and normalized ROS and gene expression levels. In sum, our results suggest the involvement of SR-BI in the maternal provision of embryonic vitamin E to the mouse embryo during neural tube closure.


Assuntos
Antígenos CD36/deficiência , Desenvolvimento Embrionário , Tubo Neural/embriologia , Tubo Neural/metabolismo , Vitamina E/metabolismo , Animais , Biomarcadores , Suplementos Nutricionais , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Oxirredução , Saco Vitelino/embriologia , Saco Vitelino/metabolismo , alfa-Tocoferol/administração & dosagem
5.
Congenit Anom (Kyoto) ; 57(5): 138-141, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28603928

RESUMO

Folate functions as a coenzyme to transfer one-carbon units that are necessary for deoxythymidylate synthesis, purine synthesis, and various methylation reactions. Ingested folate becomes a functional molecule through intestinal absorption, circulation, transport to cells, and various modifications to its structure. Associations between nutritional folate status and chronic diseases such as cardiovascular disease, cancer, and cognitive dysfunction have been reported. It has also been reported that maternal folate nutritional status is related to the risk of neural tube defects (NTDs) in the offspring. It has also been recommended that folate be consumed in the diet to promote the maintenance of good health. To reduce the risk of NTDs, supplementation with folic acid (a synthetic form of folate) during the periconceptional period has also been recommended. This paper describes the basic features and nutritional role of folate.


Assuntos
Suplementos Nutricionais , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/metabolismo , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/metabolismo , Embrião de Mamíferos , Feminino , Feto , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Humanos , Redes e Vias Metabólicas/fisiologia , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/embriologia , Defeitos do Tubo Neural/diagnóstico , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Recomendações Nutricionais
6.
Development ; 144(8): 1518-1530, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28255006

RESUMO

Folate supplementation prevents up to 70% of neural tube defects (NTDs), which result from a failure of neural tube closure during embryogenesis. The elucidation of the mechanisms underlying folate action has been challenging. This study introduces Xenopus laevis as a model to determine the cellular and molecular mechanisms involved in folate action during neural tube formation. We show that knockdown of folate receptor 1 (Folr1; also known as FRα) impairs neural tube formation and leads to NTDs. Folr1 knockdown in neural plate cells only is necessary and sufficient to induce NTDs. Folr1-deficient neural plate cells fail to constrict, resulting in widening of the neural plate midline and defective neural tube closure. Pharmacological inhibition of folate action by methotrexate during neurulation induces NTDs by inhibiting folate interaction with its uptake systems. Our findings support a model in which the folate receptor interacts with cell adhesion molecules, thus regulating the apical cell membrane remodeling and cytoskeletal dynamics necessary for neural plate folding. Further studies in this organism could unveil novel cellular and molecular events mediated by folate and lead to new ways of preventing NTDs.


Assuntos
Polaridade Celular , Receptor 1 de Folato/metabolismo , Placa Neural/citologia , Placa Neural/metabolismo , Tubo Neural/citologia , Tubo Neural/embriologia , Organogênese , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Caderinas/metabolismo , Polaridade Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Endocitose/efeitos dos fármacos , Feminino , Receptor 1 de Folato/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Marcação de Genes , Humanos , Morfolinos/farmacologia , Tubo Neural/metabolismo , Neurulação/efeitos dos fármacos , Organogênese/efeitos dos fármacos , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo
7.
Congenit Anom (Kyoto) ; 57(5): 130-133, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28244241

RESUMO

Neural tube defects (NTD) are among the most common human congenital malformations, affecting 0.5-8.0/1000 of live births. Human clinical trials have shown that periconceptional folate supplementation significantly decreases the occurrence of NTD in offspring. However, the mechanism by which folate acts on NTD remains largely unknown. Folate receptor (Folr) is one of the three membrane proteins that mediate cellular uptake of folates. Recent studies suggest that mouse Folr1 (formerly referred to as Fbp1) is essential for neural tube closure. Therefore, we examined spatial and temporal expression patterns of Folr1 in developing mouse embryos, showing a close association between Folr1 and anterior neural tube closure. Transient transgenic analysis was performed using lacZ as a reporter; we identified a 1.1-kb enhancer that directs lacZ expression in the neural tube and optic vesicle in a manner that is similar to endogenous Folr1. The 1.1-kb enhancer sequences were highly conserved between humans and mice, suggesting that human FOLR1 is associated with anterior neural tube closure in humans. Several experimental studies in mice and human epidemiological and genetics studies have suggested that folate receptor abnormalities are involved in a portion of human NTDs, although the solo defect of FOLR1 did not cause NTD.


Assuntos
Receptor 1 de Folato/genética , Ácido Fólico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Defeitos do Tubo Neural/genética , Tubo Neural/metabolismo , Animais , Embrião de Mamíferos , Elementos Facilitadores Genéticos , Feminino , Receptor 1 de Folato/metabolismo , Genes Reporter , Humanos , Óperon Lac , Camundongos , Camundongos Transgênicos , Tubo Neural/anormalidades , Tubo Neural/embriologia , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia
8.
Birth Defects Res ; 109(2): 81-91, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28008752

RESUMO

BACKGROUND: Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS: We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS: High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION: Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Proteínas de Transporte de Cátions/genética , Suplementos Nutricionais , Deficiência de Ácido Fólico/dietoterapia , Ácido Fólico/administração & dosagem , Ferro/administração & dosagem , Defeitos do Tubo Neural/prevenção & controle , Animais , Proteínas de Transporte de Cátions/deficiência , Cruzamentos Genéticos , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Deleção de Genes , Humanos , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Transgênicos , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Penetrância , Gravidez , Prosencéfalo/anormalidades , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo
9.
Birth Defects Res ; 109(2): 68-80, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-27324558

RESUMO

Susceptibility to neural tube defects (NTDs), such as anencephaly and spina bifida is influenced by genetic and environmental factors including maternal nutrition. Maternal periconceptional supplementation with folic acid significantly reduces the risk of an NTD-affected pregnancy, but does not prevent all NTDs, and "folic acid non-responsive" NTDs continue to occur. Similarly, among mouse models of NTDs, some are responsive to folic acid but others are not. Among nutritional factors, inositol deficiency causes cranial NTDs in mice while supplemental inositol prevents spinal and cranial NTDs in the curly tail (Grhl3 hypomorph) mouse, rodent models of hyperglycemia or induced diabetes, and in a folate-deficiency induced NTD model. NTDs also occur in mice lacking expression of certain inositol kinases. Inositol-containing phospholipids (phosphoinositides) and soluble inositol phosphates mediate a range of functions, including intracellular signaling, interaction with cytoskeletal proteins, and regulation of membrane identity in trafficking and cell division. Myo-inositol has been trialed in humans for a range of conditions and appears safe for use in human pregnancy. In pilot studies in Italy and the United Kingdom, women took inositol together with folic acid preconceptionally, after one or more previous NTD-affected pregnancies. In nonrandomized cohorts and a randomized double-blind study in the United Kingdom, no recurrent NTDs were observed among 52 pregnancies reported to date. Larger-scale fully powered trials are needed to determine whether supplementation with inositol and folic acid would more effectively prevent NTDs than folic acid alone. Birth Defects Research 109:68-80, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Suplementos Nutricionais , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/administração & dosagem , Inositol/administração & dosagem , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/efeitos dos fármacos , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/patologia , Humanos , Fosfatos de Inositol/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Tubo Neural/anormalidades , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Fosfatidilinositóis/metabolismo , Gravidez
10.
Birth Defects Res ; 109(1): 16-26, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-27883261

RESUMO

BACKGROUND: Neural tube defects (NTDs) are among the most common structural birth defects in humans and are caused by the complex interaction of genetic and environmental factors. Periconceptional supplementation with folic acid can prevent NTDs in both mouse models and human populations. A better understanding of how genes and environmental factors interact is critical toward development of rational strategies to prevent NTDs. Low density lipoprotein-related protein 2 (Lrp2) is involved in endocytosis of the folic acid receptor among numerous other nutrients and ligands. METHODS: We determined the effect of iron and/or folic acid supplementation on the penetrance of NTDs in the Lrp2null mouse model. The effects of supplementation on folate and iron status were measured in embryos and dams. RESULTS: Periconceptional dietary supplementation with folic acid did not prevent NTDs in Lrp2 mutant embryos, whereas high levels of folic acid supplementation by intraperitoneal injection reduced incidence of NTDs. Importantly, Lrp2null/+ dams had reduced blood folate levels that improved with daily intraperitoneal injections of folate but not dietary supplementation. On the contrary, iron supplementation had no effect on the penetrance of NTDs in Lrp2 mutant embryos and negated the preventative effect of folic acid supplementation in Lrp2null/null mutants. CONCLUSION: Lrp2 is required for folate homeostasis in heterozygous dams and high levels of supplementation prevents NTDs. Furthermore, high levels of dietary iron supplementation interfered with folic acid supplementation negating the positive effects of supplementation in this model. Birth Defects Research 109:16-26, 2017. © 2016 The Authors Birth Defects Published by Wiley Periodicals, Inc.


Assuntos
Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/prevenção & controle , Animais , Suplementos Nutricionais , Modelos Animais de Doenças , Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico , Deficiência de Ácido Fólico/complicações , Ferro/metabolismo , Proteínas Relacionadas a Receptor de LDL , Lipoproteínas , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Knockout , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética
11.
Birth Defects Res ; 109(2): 106-119, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-27860192

RESUMO

BACKGROUND: Valproic acid (VPA) is prescribed therapeutically for multiple conditions, including epilepsy. When taken during pregnancy, VPA is teratogenic, increasing the risk of several birth and developmental defects including neural tube defects (NTDs). The mechanism by which VPA causes NTDs remains controversial and how VPA interacts with folic acid (FA), a vitamin commonly recommended for the prevention of NTDs, remains uncertain. We sought to address both questions by applying untargeted metabolite profiling analysis to neural tube closure (NTC) stage mouse embryos. METHODS: Pregnant SWV dams on either a 2 ppm or 10 ppm FA supplemented diet were injected with a single dose of VPA on gestational day E8.5. On day E9.5, the mouse embryos were collected and evaluated for NTC status. Liquid chromatography coupled to mass spectrometry metabolomics analysis was performed to compare metabolite profiles of NTD-affected VPA-exposed whole mouse embryos with profiles from embryos that underwent normal NTC from control dams. RESULTS: NTDs were observed in all embryos from VPA-treated dams and penetrance was not diminished by dietary FA supplementation. The most profound metabolic perturbations were found in the 10ppm FA VPA-exposed mouse embryos, compared with the other three treatment groups. Affected metabolites included amino acids, nucleobases and related phosphorylated nucleotides, lipids, and carnitines. CONCLUSION: Maternal VPA treatment markedly perturbed purine and pyrimidine metabolism in E9.5 embryos. In combination with a high FA diet, VPA treatment resulted in gross metabolic changes, likely caused by a multiplicity of mechanisms, including an apparent disruption of mitochondrial beta-oxidation. Birth Defects Research 109:106-119, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Suplementos Nutricionais , Ácido Fólico/administração & dosagem , Defeitos do Tubo Neural/metabolismo , Neurulação/efeitos dos fármacos , Teratogênicos/toxicidade , Ácido Valproico/toxicidade , Aminoácidos/metabolismo , Animais , Carnitina/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Lipídeos/análise , Masculino , Metaboloma , Camundongos , Tubo Neural/anormalidades , Tubo Neural/efeitos dos fármacos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/patologia , Penetrância , Gravidez , Purinas/metabolismo , Pirimidinas/metabolismo
12.
Biochem Biophys Res Commun ; 467(2): 179-84, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-26453010

RESUMO

Maternal diabetes-induced birth defects remain a significant health problem. Studying the effect of natural compounds with antioxidant properties and minimal toxicities on diabetic embryopathy may lead to the development of new and safe dietary supplements. Punicalagin is a primary polyphenol found in pomegranate juice, which possesses antioxidant, anti-inflammatory and anti-tumorigenic properties, suggesting a protective effect of punicalagin on diabetic embryopathy. Here, we examined whether punicalagin could reduce high glucose-induced neural tube defects (NTDs), and if this rescue occurs through blockage of cellular stress and caspase activation. Embryonic day 8.5 (E8.5) mouse embryos were cultured for 24 or 36 h with normal (5 mM) glucose or high glucose (16.7 mM), in presence or absence of 10 or 20 µM punicalagin. 10 µM punicalagin slightly reduced NTD formation under high glucose conditions; however, 20 µM punicalagin significantly inhibited high glucose-induced NTD formation. Punicalagin suppressed high glucose-induced lipid peroxidation marker 4-hydroxynonenal, nitrotyrosine-modified proteins, and lipid peroxides. Moreover, punicalagin abrogated endoplasmic reticulum stress by inhibiting phosphorylated protein kinase ribonucleic acid (RNA)-like ER kinase (p-PERK), phosphorylated inositol-requiring protein-1α (p-IRE1α), phosphorylated eukaryotic initiation factor 2α (p-eIF2α), C/EBP-homologous protein (CHOP), binding immunoglobulin protein (BiP) and x-box binding protein 1 (XBP1) mRNA splicing. Additionally, punicalagin suppressed high glucose-induced caspase 3 and caspase 8 cleavage. Punicalagin reduces high glucose-induced NTD formation by blocking cellular stress and caspase activation. These observations suggest punicalagin supplements could mitigate the teratogenic effects of hyperglycemia in the developing embryo, and possibly prevent diabetes-induced NTDs.


Assuntos
Antioxidantes/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Taninos Hidrolisáveis/farmacologia , Defeitos do Tubo Neural/prevenção & controle , Tubo Neural/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Aldeídos/metabolismo , Animais , Caspases/genética , Caspases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Endorribonucleases/genética , Endorribonucleases/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Regulação da Expressão Gênica , Glucose , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/patologia , Fosforilação , Gravidez , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição de Fator Regulador X , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
13.
Dev Biol ; 402(1): 61-71, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25796573

RESUMO

Klhl31 is a member of the Kelch-like family in vertebrates, which are characterized by an amino-terminal broad complex tram-track, bric-a-brac/poxvirus and zinc finger (BTB/POZ) domain, carboxy-terminal Kelch repeats and a central linker region (Back domain). In developing somites Klhl31 is highly expressed in the myotome downstream of myogenic regulators (MRF), and it remains expressed in differentiated skeletal muscle. In vivo gain- and loss-of-function approaches in chick embryos reveal a role of Klhl31 in skeletal myogenesis. Targeted mis-expression of Klhl31 led to a reduced size of dermomyotome and myotome as indicated by detection of relevant myogenic markers, Pax3, Myf5, myogenin and myosin heavy chain (MF20). The knock-down of Klhl31 in developing somites, using antisense morpholinos (MO), led to an expansion of Pax3, Myf5, MyoD and myogenin expression domains and an increase in the number of mitotic cells in the dermomyotome and myotome. The mechanism underlying this phenotype was examined using complementary approaches, which show that Klhl31 interferes with ß-catenin dependent Wnt signaling. Klhl31 reduced the Wnt-mediated activation of a luciferase reporter in cultured cells. Furthermore, Klhl31 attenuated secondary axis formation in Xenopus embryos in response to Wnt1 or ß-catenin. Klhl31 mis-expression in the developing neural tube affected its dorso-ventral patterning and led to reduced dermomyotome and myotome size. Co-transfection of a Wnt3a expression vector with Klhl31 in somites or in the neural tube rescued the phenotype and restored the size of dermomyotome and myotome. Thus, Klhl31 is a novel modulator of canonical Wnt signaling, important for vertebrate myogenesis. We propose that Klhl31 acts in the myotome to support cell cycle withdrawal and differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Proliferação de Células , Embrião de Galinha , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Hibridização In Situ , Mitose , Músculos/embriologia , Miogenina/biossíntese , Tubo Neural/metabolismo , Fenótipo , Transdução de Sinais , Somitos/metabolismo , Xenopus laevis , beta Catenina/genética
14.
Birth Defects Res A Clin Mol Teratol ; 100(8): 623-32, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25115437

RESUMO

BACKGROUND: Neural tube closure defects (NTDs) are among the most common congenital malformation in human, typically presenting in liveborns as spina bifida. At least 240 gene mutations in mouse are known to increase the risk of NTD. There is a growing appreciation that environmental factors significantly contribute to NTD expression, and that NTDs likely arise from complex gene-environment interactions. Because maternal folic acid supplementation reduces human NTD risk in some populations by 60 to 70%, it is likely that NTD predisposition is often associated with a defect in folate-dependent one-carbon metabolism. A comprehensive, untargeted metabolic survey of NTD-associated changes in embryo metabolism would provide a valuable test of this assumption. We sought to establish a metabolic profiling platform that is capable of broadly assessing metabolic aberrations associated with NTD-promoting gene mutations in early-stage mouse embryos. METHODS: A liquid chromatography/mass spectrometry-based untargeted metabolite profiling platform was used to broadly identify significant differences in small molecule levels (50-1000 Da) in NTD-affected embryonic day (E) 9.5 mouse embryos (Lrp6(-) (/) (-) ) versus unaffected (Lrp6(+/+) ) control embryos. RESULTS: Results provide proof-of-principal feasibility for the broad survey of the metabolome of individual E9.5 mouse embryos and identification of metabolic changes associated with NTDs and gene mutations. Levels of 30 different metabolites were altered in association with Lrp6 gene deletion. Some metabolites link to folate-dependent one-carbon transfer reactions, as anticipated, while others await structure elucidation and pathway integration. CONCLUSION: Whole-embryo metabolomics offers the potential to identify metabolic changes in genetically determined NTD-prone embryos.


Assuntos
Ácido Fólico/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Defeitos do Tubo Neural/embriologia , Tubo Neural/embriologia , Disrafismo Espinal/embriologia , Animais , Modelos Animais de Doenças , Glutationa/metabolismo , Metaboloma/genética , Camundongos , Camundongos Knockout , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Oxirredução , Estresse Oxidativo/genética , Disrafismo Espinal/genética
15.
Birth Defects Res A Clin Mol Teratol ; 94(1): 16-21, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21954174

RESUMO

BACKGROUND: During early embryonic development, maternal exposure to hyperthermia induces neural tube defects (NTDs). Connexins are essential for the formation of gap junctions and Connexin43 (Cx43) is crucially involved in neural tube development. This study was designed to explore the potential role of Cx43 in NTDs induced by hyperthermia. METHODS: Using PCR, the Cx43 cDNA was screened from the cDNA library of the neural tube from golden hamsters treated with hyperthermia. By Northern blot, the expression of Cx43 in heat-treated and control groups of the golden hamsters at day 8.5 after mating was detected. Finally, by in situ hybridization and RT-PCR, the expression of Cx43 was examined in the neural tube at different time points after heat treatment. RESULTS: Cx43 was stably expressed in heat-treated and control groups of the golden hamsters, whereas the expression was evidently higher in the heat-treated group. Cx43 expression in the neural tube at different time points after heat treatment was significantly higher than in control groups (p < 0.01). Hyperthermia did not induce any mutations in Cx43 cDNA. CONCLUSIONS: Our data provide the first evidence that hyperthermia induces upregulation of Cx43 in the golden hamster neural tube. NTDs caused by hyperthermia may be intimately related with the overexpression of Cx43.


Assuntos
Conexina 43/metabolismo , Hipertermia Induzida/efeitos adversos , Defeitos do Tubo Neural/etiologia , Tubo Neural/metabolismo , Regulação para Cima , Animais , Conexina 43/genética , Cricetinae , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Mesocricetus , Tubo Neural/embriologia , Tubo Neural/patologia , Defeitos do Tubo Neural/genética , Reação em Cadeia da Polimerase/métodos , Gravidez , Complicações na Gravidez
16.
Genesis ; 48(5): 282-94, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20229516

RESUMO

Neural tube defects (NTDs) are the second most common birth defects in the United States. It is well known that folic acid supplementation decreases about 70% of all NTDs, although the mechanism by which this occurs is still relatively unknown. The current theory is that folic acid deficiency ultimately leads to depletion of the methyl pool, leaving critical genes unmethylated, and, in turn, their improper expression leads to failure of normal neural tube development. Recently, new studies in human cell lines have shown that folic acid deficiency and DNA hypomethylation can lead to misexpression of microRNAs (miRNAs). Misexpression of critical miRNAs during neural development may lead to a subtle effect on neural gene regulation, causing the sometimes mild to severely debilitating range of phenotypes exhibited in NTDs. This review seeks to cohesively integrate current information regarding folic acid deficiency, methylation cycles, neural development, and miRNAs to propose a potential model of NTD formation. In addition, we have examined the relevant gene pathways and miRNAs that are predicted to affect them, and based on our investigation, we have devised a basic template of experiments for exploring the idea that miRNA misregulation may be linked to folic acid deficiency and NTDs.


Assuntos
Deficiência de Ácido Fólico/complicações , MicroRNAs/metabolismo , Defeitos do Tubo Neural/metabolismo , Tubo Neural/metabolismo , Animais , Metilação de DNA , Histonas/metabolismo , Humanos , Metilação , MicroRNAs/genética , Modelos Biológicos , Tubo Neural/patologia , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/genética
17.
Hum Mol Genet ; 18(22): 4367-75, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19692351

RESUMO

The syndrome of hypomagnesemia with secondary hypocalcemia is caused by defective TRPM6. This protein is an ion channel that also contains a kinase in its C-terminus. It is usually diagnosed in childhood and, without treatment with supplemental Mg, affected children suffer from mental retardation, seizures and retarded development. We developed a mouse lacking Trpm6 in order to understand in greater detail the function of this protein. In contrast to our expectations, Trpm6(-/-) mice almost never survived to weaning. Many mice died by embryonic day 12.5. Most that survived to term had neural tube defects consisting of both exencephaly and spina bifida occulta, an unusual combination. Feeding dams a high Mg diet marginally improved offspring survival to weaning. The few Trpm6(-/-) mice that survived were fertile but matings between Trpm6(-/-) mice produced no viable pregnancies. Trpm6(+/-) mice had normal electrolytes except for modestly low plasma [Mg]. In addition, some Trpm6(+/-) mice died prematurely. Absence of Trpm6 produces an apparently different phenotype in mice than in humans. The presence of neural tube defects identifies a previously unsuspected role of Trpm6 in effecting neural tube closure. This genetic defect produces one of very few mouse models of spina bifida occulta. These results point to a critical role of Trpm6 in development and suggest an important role in neural tube closure.


Assuntos
Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/mortalidade , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Magnésio/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo
18.
Curr Med Chem ; 16(18): 2345-54, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19519395

RESUMO

Maternal diabetes develops in 2-6% of total pregnancies, depending on geographical and ethnic background. About 10% of fetuses from diabetic pregnancy display congenital malformations in various organ systems including cardiovascular, gastrointestinal, genitourinary and neurological systems, among which the neural tube defects (NTDs) such as anencephaly, holoprosencephaly and syntelencephaly were more frequently demonstrated. Recent studies by the Diabetes Control and Complications Trial Research Group show that tight glycemic control early in pregnancy decreases the progression of a number of diabetic complications. However, it appears that the pre-existing tissue damage cannot be reversed even after normoglycemic levels are achieved during pregnancy. In recent years, considerable efforts have been made to investigate the etiology of birth defects among infants of diabetic mothers. It has been shown that diabetes-induced fetal abnormalities are accompanied by some metabolic disturbances including elevated superoxide dismutase (SOD) activity, reduced levels of myoinositol and arachidonic acid and inhibition of the pentose phosphate shunt pathway. Moreover, the frequency of fetal malformations in diabetic pregnancy has been reported to be markedly reduced by dietary supplements of antioxidants such as vitamin E, vitamin C and butylated hy- droxytoluene, suggesting that oxidative stress is involved in the etiology of fetal dysmorphogenesis. Furthermore, several experimental studies have shown that NTDs in embryos of diabetic mice are associated with altered expression of genes, which control development of the neural tube. In this review, recent findings of possible molecular mechanisms which cause morphological changes during neural tube development in embryos of diabetic pregnancy are discussed.


Assuntos
Diabetes Gestacional/metabolismo , Defeitos do Tubo Neural/etiologia , Gravidez em Diabéticas/metabolismo , Animais , Apoptose , Encéfalo/embriologia , Encéfalo/metabolismo , Encéfalo/patologia , Diabetes Gestacional/epidemiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Tubo Neural/embriologia , Tubo Neural/metabolismo , Tubo Neural/patologia , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/genética , Estresse Oxidativo , Gravidez , Gravidez em Diabéticas/epidemiologia
19.
Birth Defects Res A Clin Mol Teratol ; 85(4): 295-302, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19301298

RESUMO

This review provides a brief description of folate use and folic acid metabolism in relation to neural tube defect (NTD) risk. First, a meta-analysis of reduction in NTD recurrence and occurrence risk with periconceptional folic acid supplementation is presented. Second, an overview of the complex folate metabolism is given. Third, SNPs for genes involved in folate and homocysteine metabolism that have been studied in relation to NTD riskare discussed. Fourth, the questions whether folate receptor autoantibodies or hampered methylation are mechanisms underlying NTDs are briefly discussed.


Assuntos
Ácido Fólico/metabolismo , Metiltransferases/metabolismo , Tubo Neural/embriologia , Feminino , Ácido Fólico/uso terapêutico , Variação Genética/fisiologia , Homocisteína/metabolismo , Humanos , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/fisiologia , Metilação , Modelos Biológicos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/prevenção & controle , Gravidez
20.
Neural Dev ; 2: 25, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17999760

RESUMO

BACKGROUND: The developing vertebrate brain is patterned first by global signalling gradients that define crude anteroposterior and dorsoventral coordinates, and subsequently by local signalling centres (organisers) that refine cell fate assignment within pre-patterned regions. The interface between the prethalamus and the thalamus, the zona limitans intrathalamica (ZLI), is one such local signalling centre that is essential for the establishment of these major diencephalic subdivisions by secreting the signalling factor Sonic hedgehog. Various models for ZLI formation have been proposed, but a thorough understanding of how this important local organiser is established is lacking. RESULTS: Here, we describe tissue explant experiments in chick embryos aimed at characterising the roles of different forebrain areas in ZLI formation. We found that: the ZLI becomes specified unexpectedly early; flanking regions are required for its characteristic morphogenesis; ZLI induction can occur independently from ventral tissues; interaction between any prechordal and epichordal neuroepithelial tissue anterior to the midbrain-hindbrain boundary is able to generate a ZLI; and signals from the dorsal diencephalon antagonise ZLI formation. We further show that a localised source of retinoic acid in the dorsal diencephalon is a likely candidate to mediate this inhibitory signal. CONCLUSION: Our results are consistent with a model where planar, rather than vertical, signals position the ZLI at early stages of neural development and they implicate retinoic acid as a novel molecular cue that determines its dorsoventral extent.


Assuntos
Padronização Corporal/fisiologia , Diencéfalo/embriologia , Diencéfalo/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Animais , Transplante de Tecido Encefálico/métodos , Embrião de Galinha , Coturnix , Diencéfalo/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Inibidores do Crescimento/metabolismo , Proteínas Hedgehog/metabolismo , Mesencéfalo/citologia , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Tubo Neural/citologia , Tubo Neural/embriologia , Tubo Neural/metabolismo , Neurônios/citologia , Técnicas de Cultura de Órgãos , Rombencéfalo/citologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Tálamo/citologia , Tálamo/embriologia , Tálamo/metabolismo , Quimeras de Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA