Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(11): e23738, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38855924

RESUMEN

Maternal nutrition contributes to gene-environment interactions that influence susceptibility to common congenital anomalies such as neural tube defects (NTDs). Supplemental myo-inositol (MI) can prevent NTDs in some mouse models and shows potential for prevention of human NTDs. We investigated effects of maternal MI intake on embryonic MI status and metabolism in curly tail mice, which are genetically predisposed to NTDs that are inositol-responsive but folic acid resistant. Dietary MI deficiency caused diminished MI in maternal plasma and embryos, showing that de novo synthesis is insufficient to maintain MI levels in either adult or embryonic mice. Under normal maternal dietary conditions, curly tail embryos that developed cranial NTDs had significantly lower MI content than unaffected embryos, revealing an association between diminished MI status and failure of cranial neurulation. Expression of inositol-3-phosphate synthase 1, required for inositol biosynthesis, was less abundant in the cranial neural tube than at other axial levels. Supplemental MI or d-chiro-inositol (DCI) have previously been found to prevent NTDs in curly tail embryos. Here, we investigated the metabolic effects of MI and DCI treatments by mass spectrometry-based metabolome analysis. Among inositol-responsive metabolites, we noted a disproportionate effect on nucleotides, especially purines. We also found altered proportions of 5-methyltetrahydrolate and tetrahydrofolate in MI-treated embryos suggesting altered folate metabolism. Treatment with nucleotides or the one-carbon donor formate has also been found to prevent NTDs in curly tail embryos. Together, these findings suggest that the protective effect of inositol may be mediated through the enhanced supply of nucleotides during neural tube closure.


Asunto(s)
Inositol , Defectos del Tubo Neural , Inositol/metabolismo , Inositol/farmacología , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/prevención & control , Animales , Femenino , Ratones , Embarazo , Embrión de Mamíferos/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Metaboloma , Ácido Fólico/metabolismo
2.
Genesis ; 59(11): e23445, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34490995

RESUMEN

Mouse models provide opportunities to investigate genetic interactions that cause or modify the frequency of neural tube defects (NTDs). Mutation of the PAX3 transcription factor prevents neural tube closure, leading to cranial and spinal NTDs whose frequency is responsive to folate status. Canonical Wnt signalling is implicated both in regulation of Pax3 expression and as a target of PAX3. This study investigated potential interactions of Pax3 mutation and canonical Wnt signalling using conditional gain- and loss-of-function models of ß-catenin. We found an additive effect of ß-catenin gain of function and Pax3 loss of function on NTDs and neural crest defects. ß-catenin gain of function in the Pax3 expression domain led to significantly increased frequency of cranial but not spinal NTDs in embryos that are heterozygous for Pax3 mutation, while both cranial and spinal neural tube closure were exacerbated in Pax3 homozygotes. Similarly, deficits of migrating neural crest cells were exacerbated by ß-catenin gain of function, with almost complete ablation of spinal neural crest cells and derivatives in Pax3 homozygous mutants. Pax3 expression was not affected by ß-catenin gain of function, while we confirmed that loss of function led to reduced Pax3 transcription. In contrast to gain of function, ß-catenin knockout in the Pax3 expression domain lowered the frequency of cranial NTDs in Pax3 null embryos. However, loss of function of ß-catenin and Pax3 resulted in spinal NTDs, suggesting differential regulation of cranial and spinal neural tube closure. In summary, ß-catenin function modulates the frequency of PAX3-related NTDs in the mouse.


Asunto(s)
Cresta Neural/metabolismo , Defectos del Tubo Neural/genética , Factor de Transcripción PAX3/genética , Vía de Señalización Wnt , Animales , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Mutación , Cresta Neural/anomalías , Cresta Neural/embriología , Factor de Transcripción PAX3/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Development ; 145(9)2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29636380

RESUMEN

The last stage of neural tube (NT) formation involves closure of the caudal neural plate (NP), an embryonic structure formed by neuromesodermal progenitors and newly differentiated cells that becomes incorporated into the NT. Here, we show in mouse that, as cell specification progresses, neuromesodermal progenitors and their progeny undergo significant changes in shape prior to their incorporation into the NT. The caudo-rostral progression towards differentiation is coupled to a gradual reliance on a unique combination of complex mechanisms that drive tissue folding, involving pulses of apical actomyosin contraction and planar polarised cell rearrangements, all of which are regulated by the Wnt-PCP pathway. Indeed, when this pathway is disrupted, either chemically or genetically, the polarisation and morphology of cells within the entire caudal NP is disturbed, producing delays in NT closure. The most severe disruptions of this pathway prevent caudal NT closure and result in spina bifida. In addition, a decrease in Vangl2 gene dosage also appears to promote more rapid progression towards a neural fate, but not the specification of more neural cells.


Asunto(s)
Diferenciación Celular , Placa Neural/embriología , Células-Madre Neurales/metabolismo , Tubo Neural/embriología , Vía de Señalización Wnt , Animales , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Placa Neural/patología , Células-Madre Neurales/patología , Tubo Neural/patología , Disrafia Espinal/epidemiología , Disrafia Espinal/genética , Disrafia Espinal/patología
4.
Hum Mol Genet ; 27(24): 4218-4230, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30189017

RESUMEN

The genetic basis of human neural tube defects (NTDs), such as anencephaly and spina bifida (SB), is complex and heterogeneous. Grainyhead-like genes represent candidates for involvement in NTDs based on the presence of SB and exencephaly in mice carrying loss-of-function alleles of Grhl2 or Grhl3. We found that reinstatement of Grhl3 expression, by bacterial artificial chromosome (BAC)-mediated transgenesis, prevents SB in Grhl3-null embryos, as in the Grhl3 hypomorphic curly tail strain. Notably, however, further increase in expression of Grhl3 causes highly penetrant SB. Grhl3 overexpression recapitulates the spinal NTD phenotype of loss-of-function embryos, although the underlying mechanism differs. However, it does not phenocopy other defects of Grhl3-null embryos such as abnormal axial curvature, cranial NTDs (exencephaly) or skin barrier defects, the latter being rescued by the Grhl3-transgene. Grhl2 and Grhl3 can form homodimers and heterodimers, suggesting a possible model in which defects arising from overexpression of Grhl3 result from sequestration of Grhl2 in heterodimers, mimicking Grhl2 loss of function. This hypothesis predicts that increased abundance of Grhl2 would have an ameliorating effect in Grhl3 overexpressing embryo. Instead, we observed a striking additive genetic interaction between Grhl2 and Grhl3 gain-of-function alleles. Severe SB arose in embryos in which both genes were expressed at moderately elevated levels that individually do not cause NTDs. Furthermore, moderate Grhl3 overexpression also interacted with the Vangl2Lp allele to cause SB, demonstrating genetic interaction with the planar cell polarity signalling pathway that is implicated in mouse and human NTDs.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas del Tejido Nervioso/genética , Defectos del Tubo Neural/genética , Disrafia Espinal/genética , Factores de Transcripción/genética , Alelos , Animales , Animales Modificados Genéticamente/genética , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Mutación con Pérdida de Función , Ratones , Defectos del Tubo Neural/patología , Multimerización de Proteína/genética , Disrafia Espinal/patología
5.
J Inherit Metab Dis ; 43(6): 1186-1198, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32743799

RESUMEN

Glycine abundance is modulated in a tissue-specific manner by use in biosynthetic reactions, catabolism by the glycine cleavage system (GCS), and excretion via glycine conjugation. Dysregulation of glycine metabolism is associated with multiple disorders including epilepsy, developmental delay, and birth defects. Mutation of the GCS component glycine decarboxylase (GLDC) in non-ketotic hyperglycinemia (NKH) causes accumulation of glycine in body fluids, but there is a gap in our knowledge regarding the effects on glycine metabolism in tissues. Here, we analysed mice carrying mutations in Gldc that result in severe or mild elevations of plasma glycine and model NKH. Liver of Gldc-deficient mice accumulated glycine and numerous glycine derivatives, including multiple acylglycines, indicating increased flux through reactions mediated by enzymes including glycine-N-acyltransferase and arginine: glycine amidinotransferase. Levels of dysregulated metabolites increased with age and were normalised by liver-specific rescue of Gldc expression. Brain tissue exhibited increased abundance of glycine, as well as derivatives including guanidinoacetate, which may itself be epileptogenic. Elevation of brain tissue glycine occurred even in the presence of only mildly elevated plasma glycine in mice carrying a missense allele of Gldc. Treatment with benzoate enhanced hepatic glycine conjugation thereby lowering plasma and tissue glycine. Moreover, administration of a glycine conjugation pathway intermediate, cinnamate, similarly achieved normalisation of liver glycine derivatives and circulating glycine. Although exogenous benzoate and cinnamate impact glycine levels via activity of glycine-N-acyltransferase, that is not expressed in brain, they are sufficient to lower levels of glycine and derivatives in brain tissue of treated Gldc-deficient mice.


Asunto(s)
Encéfalo/metabolismo , Glicina-Deshidrogenasa (Descarboxilante)/genética , Glicina/metabolismo , Hiperglicinemia no Cetósica/enzimología , Alelos , Animales , Encéfalo/patología , Hiperglicinemia no Cetósica/patología , Ratones , Mutación Missense
6.
Proc Natl Acad Sci U S A ; 114(26): E5177-E5186, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607062

RESUMEN

Neural tube (NT) formation in the spinal region of the mammalian embryo involves a wave of "zippering" that passes down the elongating spinal axis, uniting the neural fold tips in the dorsal midline. Failure of this closure process leads to open spina bifida, a common cause of severe neurologic disability in humans. Here, we combined a tissue-level strain-mapping workflow with laser ablation of live-imaged mouse embryos to investigate the biomechanics of mammalian spinal closure. Ablation of the zippering point at the embryonic dorsal midline causes far-reaching, rapid separation of the elevating neural folds. Strain analysis revealed tissue expansion around the zippering point after ablation, but predominant tissue constriction in the caudal and ventral neural plate zone. This zone is biomechanically coupled to the zippering point by a supracellular F-actin network, which includes an actin cable running along the neural fold tips. Pharmacologic inhibition of F-actin or laser ablation of the cable causes neural fold separation. At the most advanced somite stages, when completion of spinal closure is imminent, the cable forms a continuous ring around the neuropore, and simultaneously, a new caudal-to-rostral zippering point arises. Laser ablation of this new closure initiation point causes neural fold separation, demonstrating its biomechanical activity. Failure of spinal closure in pre-spina bifida Zic2Ku mutant embryos is associated with altered tissue biomechanics, as indicated by greater neuropore widening after ablation. Thus, this study identifies biomechanical coupling of the entire region of active spinal neurulation in the mouse embryo as a prerequisite for successful NT closure.


Asunto(s)
Embrión de Mamíferos/metabolismo , Modelos Biológicos , Tubo Neural/embriología , Actinas , Animales , Embrión de Mamíferos/citología , Humanos , Ratones , Ratones Mutantes , Tubo Neural/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Dev Biol ; 435(2): 130-137, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29397878

RESUMEN

Failure of neural tube closure leads to neural tube defects (NTDs), common congenital abnormalities in humans. Among the genes whose loss of function causes NTDs in mice, Grainyhead-like3 (Grhl3) is essential for spinal neural tube closure, with null mutants exhibiting fully penetrant spina bifida. During spinal neurulation Grhl3 is initially expressed in the surface (non-neural) ectoderm, subsequently in the neuroepithelial component of the neural folds and at the node-streak border, and finally in the hindgut endoderm. Here, we show that endoderm-specific knockout of Grhl3 causes late-arising spinal NTDs, preceded by increased ventral curvature of the caudal region which was shown previously to suppress closure of the spinal neural folds. This finding supports the hypothesis that diminished Grhl3 expression in the hindgut is the cause of spinal NTDs in the curly tail, carrying a hypomorphic Grhl3 allele. Complete loss of Grhl3 function produces a more severe phenotype in which closure fails earlier in neurulation, before the stage of onset of expression in the hindgut of wild-type embryos. This implicates additional tissues and NTD mechanisms in Grhl3 null embryos. Conditional knockout of Grhl3 in the neural plate and node-streak border has minimal effect on closure, suggesting that abnormal function of surface ectoderm, where Grhl3 transcripts are first detected, is primarily responsible for early failure of spinal neurulation in Grhl3 null embryos.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Defectos del Tubo Neural/genética , Tubo Neural/fisiología , Neurulación/genética , Factores de Transcripción/fisiología , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Estratos Germinativos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Placa Neural/metabolismo , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Especificidad de Órganos , ARN Mensajero/biosíntesis , Disrafia Espinal/embriología , Disrafia Espinal/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
8.
J Cell Sci ; 128(14): 2468-81, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26040287

RESUMEN

The cytoskeleton is widely considered essential for neurulation, yet the mouse spinal neural tube can close despite genetic and non-genetic disruption of the cytoskeleton. To investigate this apparent contradiction, we applied cytoskeletal inhibitors to mouse embryos in culture. Preventing actomyosin cross-linking, F-actin assembly or myosin II contractile activity did not disrupt spinal closure. In contrast, inhibiting Rho kinase (ROCK, for which there are two isoforms ROCK1 and ROCK2) or blocking F-actin disassembly prevented closure, with apical F-actin accumulation and adherens junction disturbance in the neuroepithelium. Cofilin-1-null embryos yielded a similar phenotype, supporting the hypothesis that there is a key role for actin turnover. Co-exposure to Blebbistatin rescued the neurulation defects caused by RhoA inhibition, whereas an inhibitor of myosin light chain kinase, ML-7, had no such effect. We conclude that regulation of RhoA, Rho kinase, LIM kinase and cofilin signalling is necessary for spinal neural tube closure through precise control of neuroepithelial actin turnover and actomyosin disassembly. In contrast, actomyosin assembly and myosin ATPase activity are not limiting for closure.


Asunto(s)
Actinas/metabolismo , Actomiosina/metabolismo , Tubo Neural/embriología , Quinasas Asociadas a rho/metabolismo , Actinas/genética , Actomiosina/genética , Animales , Cofilina 1/genética , Cofilina 1/metabolismo , Quinasas Lim/genética , Quinasas Lim/metabolismo , Ratones , Ratones Mutantes , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/genética , Proteína de Unión al GTP rhoA
9.
Development ; 141(16): 3153-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25038043

RESUMEN

The role of planar cell polarity (PCP) signalling in neural crest (NC) development is unclear. The PCP dependence of NC cell migration has been reported in Xenopus and zebrafish, but NC migration has not been studied in mammalian PCP mutants. Vangl2(Lp/Lp) mouse embryos lack PCP signalling and undergo almost complete failure of neural tube closure. Here we show, however, that NC specification, migration and derivative formation occur normally in Vangl2(Lp/Lp) embryos. The gene family member Vangl1 was not expressed in NC nor ectopically expressed in Vangl2(Lp/Lp) embryos, and doubly homozygous Vangl1/Vangl2 mutants exhibited normal NC migration. Acute downregulation of Vangl2 in the NC lineage did not prevent NC migration. In vitro, Vangl2(Lp/Lp) neural tube explants generated emigrating NC cells, as in wild type. Hence, PCP signalling is not essential for NC migration in mammals, in contrast to its essential role in neural tube closure. PCP mutations are thus unlikely to mediate NC-related birth defects in humans.


Asunto(s)
Proteínas Portadoras/fisiología , Polaridad Celular/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Cresta Neural/citología , Alelos , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Homocigoto , Proteínas de la Membrana/genética , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Cresta Neural/metabolismo , Tubo Neural/embriología , Transducción de Señal
10.
Prenat Diagn ; 37(3): 273-281, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28056489

RESUMEN

OBJECTIVE: We used non-invasive high-frequency ultrasound (HFUS) imaging to investigate embryonic brain development in a mouse model for neural tube defects (NTDs) and non-ketotic hyperglycinemia (NKH). METHOD: Using HFUS, we imaged embryos carrying loss of function alleles of Gldc encoding glycine decarboxylase, a component of the glycine cleavage system in mitochondrial folate metabolism, which is known to be associated with cranial NTDs and NKH in humans. We serially examined the same litter during the second half of embryonic development and quantified cerebral structures. Genotype was confirmed using PCR. Histology was used to confirm ultrasound findings. RESULTS: High-frequency ultrasound allowed in utero detection of two major brain abnormalities in Gldc-deficient mouse embryos, cranial NTDs (exencephaly) and ventriculomegaly (corresponding with the previous finding of post-natal hydrocephalus). Serial ultrasound allowed individual embryos to be analysed at successive gestational time points. From embryonic day 16.5 to 18.5, the lateral ventricle volume reduced in wild-type and heterozygous embryos but increased in homozygous Gldc-deficient embryos. CONCLUSION: Exencephaly and ventriculomegaly were detectable by HFUS in homozygous Gldc-deficient mouse embryos indicating this to be an effective tool to study CNS development. Longitudinal analysis of the same embryo allowed the prenatal onset and progression of ventricle enlargement in Gldc-deficient mice to be determined. © 2017 The Authors. Prenatal Diagnosis published by John Wiley & Sons, Ltd.


Asunto(s)
Glicina-Deshidrogenasa (Descarboxilante)/genética , Hidrocefalia/diagnóstico , Defectos del Tubo Neural/diagnóstico , Ultrasonografía Prenatal , Animales , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/embriología , Embrión de Mamíferos , Femenino , Hidrocefalia/embriología , Hidrocefalia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Embarazo , Cráneo/diagnóstico por imagen , Cráneo/embriología , Ultrasonografía Prenatal/métodos
11.
Dev Biol ; 404(2): 113-24, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26079577

RESUMEN

Bending of the neural plate at paired dorsolateral hinge points (DLHPs) is required for neural tube closure in the spinal region of the mouse embryo. As a step towards understanding the morphogenetic mechanism of DLHP development, we examined variations in neural plate cellular architecture and proliferation during closure. Neuroepithelial cells within the median hinge point (MHP) contain nuclei that are mainly basally located and undergo relatively slow proliferation, with a 7 h cell cycle length. In contrast, cells in the dorsolateral neuroepithelium, including the DLHP, exhibit nuclei distributed throughout the apico-basal axis and undergo rapid proliferation, with a 4 h cell cycle length. As the neural folds elevate, cell numbers increase to a greater extent in the dorsolateral neural plate that contacts the surface ectoderm, compared with the more ventromedial neural plate where cells contact paraxial mesoderm and notochord. This marked increase in dorsolateral cell number cannot be accounted for solely on the basis of enhanced cell proliferation in this region. We hypothesised that neuroepithelial cells may translocate in a ventral-to-dorsal direction as DLHP formation occurs, and this was confirmed by vital cell labelling in cultured embryos. The translocation of cells into the neural fold, together with its more rapid cell proliferation, leads to an increase in cell density dorsolaterally compared with the more ventromedial neural plate. These findings suggest a model in which DLHP formation may proceed through 'buckling' of the neuroepithelium at a dorso-ventral boundary marked by a change in cell-packing density.


Asunto(s)
Placa Neural/embriología , Tubo Neural/embriología , Células Neuroepiteliales/citología , Neurulación/fisiología , Animales , Movimiento Celular , Proliferación Celular , Ciclina D1/metabolismo , Embrión de Mamíferos , Mesodermo/citología , Ratones , Ratones Endogámicos BALB C , Cresta Neural/citología , Placa Neural/citología , Tubo Neural/citología , Notocorda/citología , Médula Espinal/citología
12.
Brain ; 136(Pt 9): 2836-41, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23935126

RESUMEN

Closure of the neural tube during embryogenesis is a crucial step in development of the central nervous system. Failure of this process results in neural tube defects, including spina bifida and anencephaly, which are among the most common birth defects worldwide. Maternal use of folic acid supplements reduces risk of neural tube defects but a proportion of cases are not preventable. Folic acid is thought to act through folate one-carbon metabolism, which transfers one-carbon units for methylation reactions and nucleotide biosynthesis. Hence suboptimal performance of the intervening reactions could limit the efficacy of folic acid. We hypothesized that direct supplementation with nucleotides, downstream of folate metabolism, has the potential to support neural tube closure. Therefore, in a mouse model that exhibits folic acid-resistant neural tube defects, we tested the effect of specific combinations of pyrimidine and purine nucleotide precursors and observed a significant protective effect. Labelling in whole embryo culture showed that nucleotides are taken up by the neurulating embryo and incorporated into genomic DNA. Furthermore, the mitotic index was elevated in neural folds and hindgut of treated embryos, consistent with a proposed mechanism of neural tube defect prevention through stimulation of cellular proliferation. These findings may provide an impetus for future investigations of supplemental nucleotides as a means to prevent a greater proportion of human neural tube defects than can be achieved by folic acid alone.


Asunto(s)
Ácido Fólico/efectos adversos , Defectos del Tubo Neural/prevención & control , Nucleósidos de Purina/uso terapéutico , Nucleósidos de Pirimidina/uso terapéutico , Animales , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/fisiología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Ácido Fólico/metabolismo , Histonas/metabolismo , Tamaño de la Camada/efectos de los fármacos , Masculino , Exposición Materna , Ratones , Ratones Mutantes , Defectos del Tubo Neural/tratamiento farmacológico , Defectos del Tubo Neural/genética , Embarazo , Estadísticas no Paramétricas , Timidina/uso terapéutico
13.
Hum Mol Genet ; 20(8): 1536-46, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21262862

RESUMEN

Cranial neural tube defects (NTDs) occur in mice carrying mutant alleles of many different genes, whereas isolated spinal NTDs (spina bifida) occur in fewer models, despite being common human birth defects. Spina bifida occurs at high frequency in the Axial defects (Axd) mouse mutant but the causative gene is not known. In the current study, the Axd mutation was mapped by linkage analysis. Within the critical genomic region, sequencing did not reveal a coding mutation whereas expression analysis demonstrated significant up-regulation of grainyhead-like 2 (Grhl2) in Axd mutant embryos. Expression of other candidate genes did not differ between genotypes. In order to test the hypothesis that over-expression of Grhl2 causes Axd NTDs, we performed a genetic cross to reduce Grhl2 function in Axd heterozygotes. Grhl2 loss of function mutant mice were generated and displayed both cranial and spinal NTDs. Compound heterozygotes carrying both loss (Grhl2 null) and putative gain of function (Axd) alleles exhibited normalization of spinal neural tube closure compared with Axd/+ littermates, which exhibit delayed closure. Grhl2 is expressed in the surface ectoderm and hindgut endoderm in the spinal region, overlapping with grainyhead-like 3 (Grhl3). Axd mutants display delayed eyelid closure, as reported in Grhl3 null embryos. Moreover, Axd mutant embryos exhibited increased ventral curvature of the spinal region and reduced proliferation in the hindgut, reminiscent of curly tail embryos, which carry a hypomorphic allele of Grhl3. Overall, our data suggest that defects in Axd mutant embryos result from over-expression of Grhl2.


Asunto(s)
Disrafia Espinal/genética , Factores de Transcripción/genética , Animales , Proliferación Celular , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Femenino , Silenciador del Gen , Ligamiento Genético , Humanos , Hibridación Genética , Tracto Gastrointestinal Inferior/anomalías , Tracto Gastrointestinal Inferior/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Disrafia Espinal/embriología , Factores de Transcripción/metabolismo , Transcripción Genética , Regulación hacia Arriba
14.
Dis Model Mech ; 16(3)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36916392

RESUMEN

Understanding the molecular mechanisms that lead to birth defects is an important step towards improved primary prevention. Mouse embryos homozygous for the Kumba (Ku) mutant allele of Zic2 develop severe spina bifida with complete lack of dorsolateral hinge points (DLHPs) in the neuroepithelium. Bone morphogenetic protein (BMP) signalling is overactivated in Zic2Ku/Ku embryos, and the BMP inhibitor dorsomorphin partially rescues neural tube closure in cultured embryos. RhoA signalling is also overactivated, with accumulation of actomyosin in the Zic2Ku/Ku neuroepithelium, and the myosin inhibitor Blebbistatin partially normalises neural tube closure. However, dorsomorphin and Blebbistatin differ in their effects at tissue and cellular levels: DLHP formation is rescued by dorsomorphin but not Blebbistatin, whereas abnormal accumulation of actomyosin is rescued by Blebbistatin but not dorsomorphin. These findings suggest a dual mechanism of spina bifida origin in Zic2Ku/Ku embryos: faulty BMP-dependent formation of DLHPs and RhoA-dependent F-actin accumulation in the neuroepithelium. Hence, we identify a multi-pathway origin of spina bifida in a mammalian system that may provide a developmental basis for understanding the corresponding multifactorial human defects.


Asunto(s)
Defectos del Tubo Neural , Disrafia Espinal , Ratones , Animales , Humanos , Tubo Neural/metabolismo , Actomiosina/metabolismo , Defectos del Tubo Neural/genética , Neurulación , Mamíferos/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
15.
Proc Natl Acad Sci U S A ; 106(20): 8233-8, 2009 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-19420217

RESUMEN

Apoptotic cell death occurs in many tissues during embryonic development and appears to be essential for processes including digit formation and cardiac outflow tract remodeling. Studies in the chick suggest a requirement for apoptosis during neurulation, because inhibition of caspase activity was found to prevent neural tube closure. In mice, excessive apoptosis occurs in association with failure of neural tube closure in several genetic mutants, but whether regulated apoptosis is also necessary for neural tube closure in mammals is unknown. Here we investigate the possible role of apoptotic cell death during mouse neural tube closure. We confirm the presence of apoptosis in the neural tube before and during closure, and identify a correlation with 3 main events: bending and fusion of the neural folds, postfusion remodeling of the dorsal neural tube and surface ectoderm, and emigration of neural crest cells. Both Casp3 and Apaf1 null embryos exhibit severely reduced apoptosis, yet neurulation proceeds normally in the forebrain and spine. In contrast, the mutant embryos fail to complete neural tube closure in the midbrain and hindbrain. Application of the apoptosis inhibitors z-Vad-fmk and pifithrin-alpha to neurulation-stage embryos in culture suppresses apoptosis but does not prevent initiation or progression of neural tube closure along the entire neuraxis, including the midbrain and hindbrain. Remodeling of the surface ectoderm to cover the closed tube, as well as delamination and migration of neural crest cells, also appear to be normal in the apoptosis-suppressed embryos. We conclude that apoptosis is not required for neural tube closure in the mouse embryo.


Asunto(s)
Apoptosis/fisiología , Tubo Neural/embriología , Animales , Factor Apoptótico 1 Activador de Proteasas , Caspasa 3 , Movimiento Celular , Ectodermo/citología , Embrión de Mamíferos , Ratones , Cresta Neural/citología , Tubo Neural/citología
16.
Sci Rep ; 12(1): 9693, 2022 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690633

RESUMEN

Children with syndromic forms of craniosynostosis undergo a plethora of surgical interventions to resolve the clinical features caused by the premature fusion of cranial sutures. While surgical correction is reliable, the need for repeated rounds of invasive treatment puts a heavy burden on the child and their family. This study explores a non-surgical alternative using mechanical loading of the cranial joints to prevent or delay craniofacial phenotypes associated with Crouzon syndrome. We treated Crouzon syndrome mice before the onset of craniosynostosis by cyclical mechanical loading of cranial joints using a custom designed set-up. Cranial loading applied to the frontal bone partially restores normal skull morphology, significantly reducing the typical brachycephalic appearance. This is underpinned by the delayed closure of the coronal suture and of the intersphenoidal synchondrosis. This study provides a novel treatment alternative for syndromic craniosynostosis which has the potential to be an important step towards replacing, reducing or refining the surgical treatment of all craniosynostosis patients.


Asunto(s)
Disostosis Craneofacial , Craneosinostosis , Animales , Suturas Craneales/cirugía , Disostosis Craneofacial/cirugía , Craneosinostosis/genética , Craneosinostosis/cirugía , Hueso Frontal , Humanos , Ratones , Fenotipo , Cráneo/cirugía
17.
Dis Model Mech ; 15(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34842271

RESUMEN

Planar cell polarity (PCP) signalling is vital for initiation of mouse neurulation, with diminished convergent extension (CE) cell movements leading to craniorachischisis, a severe neural tube defect (NTD). Some humans with NTDs also have PCP gene mutations but these are heterozygous, not homozygous as in mice. Other genetic or environmental factors may interact with partial loss of PCP function in human NTDs. We found that reduced sulfation of glycosaminoglycans interacts with heterozygosity for the Lp allele of Vangl2 (a core PCP gene), to cause craniorachischisis in cultured mouse embryos, with rescue by exogenous sulphate. We hypothesized that this glycosaminoglycan-PCP interaction may regulate CE, but, surprisingly, DiO labelling of the embryonic node demonstrates no abnormality of midline axial extension in sulfation-depleted Lp/+ embryos. Positive-control Lp/Lp embryos show severe CE defects. Abnormalities were detected in the size and shape of somites that flank the closing neural tube in sulfation-depleted Lp/+ embryos. We conclude that failure of closure initiation can arise by a mechanism other than faulty neuroepithelial CE, with possible involvement of matrix-mediated somite expansion, adjacent to the closing neural tube.


Asunto(s)
Polaridad Celular , Defectos del Tubo Neural , Animales , Interacción Gen-Ambiente , Ratones , Proteínas del Tejido Nervioso/genética , Tubo Neural , Defectos del Tubo Neural/genética
18.
Hum Mol Genet ; 17(23): 3675-85, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18753144

RESUMEN

Risk of neural tube defects (NTDs) is determined by genetic and environmental factors, among which folate status appears to play a key role. However, the precise nature of the link between low folate status and NTDs is poorly understood, and it remains unclear how folic acid prevents NTDs. We investigated the effect of folate level on risk of NTDs in splotch (Sp(2)(H)) mice, which carry a mutation in Pax3. Dietary folate restriction results in reduced maternal blood folate, elevated plasma homocysteine and reduced embryonic folate content. Folate deficiency does not cause NTDs in wild-type mice, but causes a significant increase in cranial NTDs among Sp(2)(H) embryos, demonstrating a gene-environment interaction. Control treatments, in which intermediate levels of folate are supplied, suggest that NTD risk is related to embryonic folate concentration, not maternal blood folate concentration. Notably, the effect of folate deficiency appears more deleterious in female embryos than males, since defects are not prevented by exogenous folic acid. Folate-deficient embryos exhibit developmental delay and growth retardation. However, folate content normalized to protein content is appropriate for developmental stage, suggesting that folate availability places a tight limit on growth and development. Folate-deficient embryos also exhibit a reduced ratio of s-adenosylmethionine (SAM) to s-adenosylhomocysteine (SAH). This could indicate inhibition of the methylation cycle, but we did not detect any diminution in global DNA methylation, in contrast to embryos in which the methylation cycle was specifically inhibited. Hence, folate deficiency increases the risk of NTDs in genetically predisposed splotch embryos, probably via embryonic growth retardation.


Asunto(s)
Susceptibilidad a Enfermedades/metabolismo , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Factores de Transcripción Paired Box/genética , Animales , Femenino , Ácido Fólico/sangre , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/embriología , Deficiencia de Ácido Fólico/fisiopatología , Homocisteína/sangre , Homocisteína/metabolismo , Humanos , Masculino , Metilación , Ratones , Ratones Transgénicos , Mutación , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/fisiopatología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/metabolismo
19.
Birth Defects Res A Clin Mol Teratol ; 88(8): 612-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20589880

RESUMEN

BACKGROUND: Folate one-carbon metabolism has been implicated as a determinant of susceptibility to neural tube defects (NTDs), owing to the preventive effect of maternal folic acid supplementation and the higher risk associated with markers of diminished folate status. METHODS: Folate one-carbon metabolism was compared in curly tail (ct/ct) and genetically matched congenic (+(ct)/+(ct)) mouse strains using the deoxyuridine suppression test in embryonic fibroblast cells and by quantifying s-adenosylmethionine (SAM) and s-adenosylhomocysteine (SAH) in embryos using liquid chromatography tandem mass spectrometry. A possible genetic interaction between curly tail and a null allele of 5,10-methylenetetrahydrofolate reductase (MTHFR) was investigated by generation of compound mutant embryos. RESULTS: There was no deficit in thymidylate biosynthesis in ct/ct cells, but incorporation of exogenous thymidine was lower than in +(ct)/+(ct) cells. In +(ct)/+(ct) embryos the SAM/SAH ratio was diminished by dietary folate deficiency and normalized by folic acid or myo-inositol treatment, in association with prevention of NTDs. In contrast, folate deficiency caused a significant increase in the SAM/SAH ratio in ct/ct embryos. Loss of MTHFR function in curly tail embryos significantly reduced the SAM/SAH ratio but did not cause cranial NTDs or alter the frequency of caudal NTDs. CONCLUSIONS: Curly tail fibroblasts and embryos, in which Grhl3 expression is reduced, display alterations in one-carbon metabolism, particularly in the response to folate deficiency, compared to genetically matched congenic controls in which Grhl3 is unaffected. However, unlike folate deficiency, diminished methylation potential appears to be insufficient to cause cranial NTDs in the curly tail strain, nor does it increase the frequency of caudal NTDs.


Asunto(s)
Carbono/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Ácido Fólico/metabolismo , Defectos del Tubo Neural/etiología , Animales , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/metabolismo , Deficiencia de Ácido Fólico/genética , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Ratones , Ratones Mutantes , Defectos del Tubo Neural/metabolismo , Embarazo , S-Adenosilhomocisteína/análisis , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/análisis , S-Adenosilmetionina/metabolismo , Timidina Monofosfato/biosíntesis , Factores de Transcripción/genética
20.
Birth Defects Res ; 112(2): 196-204, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31793758

RESUMEN

CreERT2-mediated gene recombination is widely applied in developmental biology research. Activation of CreERT2 is typically achieved by injection of tamoxifen in an oily vehicle into the peritoneal cavity of mid-gestation pregnant mice. This can be technically challenging and adversely impacts welfare. Here we characterize three refinements to this technique: Pipette feeding (not gavage) of tamoxifen, ex vivo CreERT2 activation in whole embryo culture and injection of cell-permeable TAT-Cre into Cre-negative cultured embryos. We demonstrate that pipette feeding of tamoxifen solution to the mother on various days of gestation reliably activates embryonic CreERT2, illustrated here using ß-Actin CreERT2 , Sox2 CreERT2 , T CreERT2 , and Nkx1.2 CreERT2 . Pipette feeding of tamoxifen induces dose-dependent recombination of Rosa26 mTmG reporters when administered at E8.5. Activation of two neuromesodermal progenitor-targeting Cre drivers, T CreERT2 , and Nkx1.2 CreERT2 , produces comparable neuroepithelial lineage tracing. Dose-dependent CreERT2 activation can also be achieved by brief exposure to 4OH-tamoxifen in whole embryo culture, allowing temporal control of gene deletion and eliminating the need to treat pregnant mice. Rosa26 mTmG reporter recombination can also be achieved regionally by injecting TAT-Cre into embryonic tissues at the start of culture. This allows greater spatial control over Cre activation than can typically be achieved with endogenous CreERT2, for example by injecting TAT-Cre on one side of the midline. We hope that our description and application of these techniques will stimulate refinement of experimental methods involving CreERT2 activation for gene deletion and lineage tracing studies. Improved temporal (ex vivo treatment) and spatial (TAT-Cre injection) control of recombination will also allow previously intractable questions to be addressed.


Asunto(s)
Regulación de la Expresión Génica/genética , Ingeniería Genética/métodos , Animales , Aberraciones Cromosómicas/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Eliminación de Gen , Expresión Génica/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Embarazo , Recombinación Genética/genética , Tamoxifeno/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA