Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Genet Metab ; 142(3): 108496, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38761651

RESUMEN

Non-Ketotic Hyperglycinemia (NKH) is a rare inborn error of metabolism caused by impaired function of the glycine cleavage system (GCS) and characterised by accumulation of glycine in body fluids and tissues. NKH is an autosomal recessive condition and the majority of affected individuals carry mutations in GLDC (glycine decarboxylase). Current treatments for NKH have limited effect and are not curative. As a monogenic condition with known genetic causation, NKH is potentially amenable to gene therapy. An AAV9-based expression vector was designed to target sites of GCS activity. Using a ubiquitous promoter to drive expression of a GFP reporter, transduction of liver and brain was confirmed following intra-venous and/or intra-cerebroventricular administration to neonatal mice. Using the same capsid and promoter with transgenes to express mouse or human GLDC, vectors were then tested in GLDC-deficient mice that provide a model of NKH. GLDC-deficient mice exhibited elevated plasma glycine concentration and accumulation of glycine in liver and brain tissues as previously observed. Moreover, the folate profile indicated suppression of folate one­carbon metabolism (FOCM) in brain tissue, as found at embryonic stages, and reduced abundance of FOCM metabolites including betaine and choline. Neonatal administration of vector achieved reinstatement of GLDC mRNA and protein expression in GLDC-deficient mice. Treated GLDC-deficient mice showed significant lowering of plasma glycine, confirming functionality of vector expressed protein. AAV9-GLDC treatment also led to lowering of brain tissue glycine, and normalisation of the folate profile indicating restoration of glycine-derived one­carbon supply. These findings support the hypothesis that AAV-mediated gene therapy may offer potential in treatment of NKH.

2.
Mol Cell Proteomics ; 23(3): 100718, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38224738

RESUMEN

A functional role has been ascribed to the human dihydrofolate reductase 2 (DHFR2) gene based on the enzymatic activity of recombinant versions of the predicted translated protein. However, the in vivo function is still unclear. The high amino acid sequence identity (92%) between DHFR2 and its parental homolog, DHFR, makes analysis of the endogenous protein challenging. This paper describes a targeted mass spectrometry proteomics approach in several human cell lines and tissue types to identify DHFR2-specific peptides as evidence of its translation. We show definitive evidence that the DHFR2 activity in the mitochondria is in fact mediated by DHFR, and not DHFR2. Analysis of Ribo-seq data and an experimental assessment of ribosome association using a sucrose cushion showed that the two main Ensembl annotated mRNA isoforms of DHFR2, 201 and 202, are differentially associated with the ribosome. This indicates a functional role at both the RNA and protein level. However, we were unable to detect DHFR2 protein at a detectable level in most cell types examined despite various RNA isoforms of DHFR2 being relatively abundant. We did detect a DHFR2-specific peptide in embryonic heart, indicating that the protein may have a specific role during embryogenesis. We propose that the main functionality of the DHFR2 gene in adult cells is likely to arise at the RNA level.


Asunto(s)
ARN , Tetrahidrofolato Deshidrogenasa , Humanos , Línea Celular , Péptidos/metabolismo , Biosíntesis de Proteínas , Ribosomas/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo
3.
Hum Mol Genet ; 32(17): 2681-2692, 2023 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-37364051

RESUMEN

Orofacial clefts, including cleft lip and palate (CL/P) and neural tube defects (NTDs) are among the most common congenital anomalies, but knowledge of the genetic basis of these conditions remains incomplete. The extent to which genetic risk factors are shared between CL/P, NTDs and related anomalies is also unclear. While identification of causative genes has largely focused on coding and loss of function mutations, it is hypothesized that regulatory mutations account for a portion of the unidentified heritability. We found that excess expression of Grainyhead-like 2 (Grhl2) causes not only spinal NTDs in Axial defects (Axd) mice but also multiple additional defects affecting the cranial region. These include orofacial clefts comprising midline cleft lip and palate and abnormalities of the craniofacial bones and frontal and/or basal encephalocele, in which brain tissue herniates through the cranium or into the nasal cavity. To investigate the causative mutation in the Grhl2Axd strain, whole genome sequencing identified an approximately 4 kb LTR retrotransposon insertion that disrupts the non-coding regulatory region, lying approximately 300 base pairs upstream of the 5' UTR. This insertion also lies within a predicted long non-coding RNA, oriented on the reverse strand, which like Grhl2 is over-expressed in Axd (Grhl2Axd) homozygous mutant embryos. Initial analysis of the GRHL2 upstream region in individuals with NTDs or cleft palate revealed rare or novel variants in a small number of cases. We hypothesize that mutations affecting the regulation of GRHL2 may contribute to craniofacial anomalies and NTDs in humans.


Asunto(s)
Anomalías Múltiples , Labio Leporino , Fisura del Paladar , Defectos del Tubo Neural , Disrafia Espinal , Animales , Humanos , Ratones , Anomalías Múltiples/genética , Labio Leporino/genética , Fisura del Paladar/genética , Encefalocele/genética , Mutación , Defectos del Tubo Neural/genética , Disrafia Espinal/genética
4.
Front Genet ; 12: 625120, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33569080

RESUMEN

Glycine cleavage system H protein (GCSH) is a component of the glycine cleavage system (GCS), a conserved protein complex that acts to decarboxylate glycine. Mutation of AMT or GLDC, encoding the GCS components aminomethyltransferase and glycine decarboxylase, can cause malformations of the developing CNS (neural tube defects (NTDs) and ventriculomegaly) as well as a post-natal life-limiting neurometabolic disorder, Non-Ketotic Hyperglycinemia. In contrast, it is unclear whether mutation of GCSH contributes to these conditions and we therefore investigated GCSH loss of function in mice. Mice that were heterozygous for a Gcsh null allele were viable and did not exhibit elevated plasma glycine. Moreover, heterozygous mutation of Gcsh did not increase the frequency of NTDs in Gldc mutant embryos. Homozygous Gcsh null mice were not recovered at post-natal stages. Analysis of litters at E8.5-10.5, revealed the presence of homozygous null embryos which were much smaller than littermates and had failed to develop beyond early post-implantation stages with no visible somites or head-folds. Hence, unlike null mutations of Gldc or Amt, which are compatible with embryonic survival despite the presence of NTDs, loss of Gcsh causes embryonic death prior to mid-gestation. Maternal supplementation with formate did not restore embryonic development beyond E7.5, suggesting that the primary cause of lethality was not loss of glycine cleavage activity or suppression of folate one-carbon metabolism. These findings suggest that GCSH has additional roles beyond function in the glycine cleavage system. We hypothesize that GCSH potentially acts in lipoylation of 2-oxoacid dehydrogenase proteins, as reported in bacteria.

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.
Birth Defects Res ; 112(2): 205-211, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31758757

RESUMEN

BACKGROUND: Neural tube defects (NTDs) result from failure of neural tube closure during embryogenesis. These severe birth defects of the central nervous system include anencephaly and spina bifida, and affect 0.5-2 per 1,000 pregnancies worldwide in humans. It has been demonstrated that acetylation plays a pivotal role during neural tube closure, as animal models for defective histone acetyltransferase proteins display NTDs. Acetylation represents an important component of the complex network of posttranslational regulatory interactions, suggesting a possible fundamental role during primary neurulation events. This study aimed to assess protein acetylation contribution to early patterning of the central nervous system both in human and murine specimens. METHODS: We used both human and mouse (Cited2 -/- ) samples to analyze the dynamic acetylation of proteins during embryo development through immunohistochemistry, western blot analysis and quantitative polymerase chain reaction. RESULTS: We report the dynamic profile of histone and protein acetylation status during neural tube closure. We also report a rescue effect in an animal model by chemical p53 inhibition. CONCLUSIONS: Our data suggest that the p53-acetylation equilibrium may play a role in primary neurulation in mammals.


Asunto(s)
Defectos del Tubo Neural/embriología , Neurulación/genética , Acetilación , Anencefalia/etiología , Anencefalia/fisiopatología , Animales , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Histona Acetiltransferasas/metabolismo , Humanos , Mamíferos , Ratones/embriología , Neurulación/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Disrafia Espinal/etiología , Disrafia Espinal/fisiopatología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
Dis Model Mech ; 12(11)2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31636139

RESUMEN

Neural tube defects (NTDs), including spina bifida and anencephaly, are among the most common birth defects worldwide, but their underlying genetic and cellular causes are not well understood. Some NTDs are preventable by supplemental folic acid. However, despite widespread use of folic acid supplements and implementation of food fortification in many countries, the protective mechanism is unclear. Pax3 mutant (splotch; Sp2H ) mice provide a model in which NTDs are preventable by folic acid and exacerbated by maternal folate deficiency. Here, we found that cell proliferation was diminished in the dorsal neuroepithelium of mutant embryos, corresponding to the region of abolished Pax3 function. This was accompanied by premature neuronal differentiation in the prospective midbrain. Contrary to previous reports, we did not find evidence that increased apoptosis could underlie failed neural tube closure in Pax3 mutant embryos, nor that inhibition of apoptosis could prevent NTDs. These findings suggest that Pax3 functions to maintain the neuroepithelium in a proliferative, undifferentiated state, allowing neurulation to proceed. NTDs in Pax3 mutants were not associated with abnormal abundance of specific folates and were not prevented by formate, a one-carbon donor to folate metabolism. Supplemental folic acid restored proliferation in the cranial neuroepithelium. This effect was mediated by enhanced progression of the cell cycle from S to G2 phase, specifically in the Pax3 mutant dorsal neuroepithelium. We propose that the cell-cycle-promoting effect of folic acid compensates for the loss of Pax3 and thereby prevents cranial NTDs.


Asunto(s)
Ácido Fólico/administración & dosificación , Mutación , Defectos del Tubo Neural/etiología , Factor de Transcripción PAX3/genética , Animales , Apoptosis , Ciclo Celular/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos CBA , Defectos del Tubo Neural/prevención & control , Factor de Transcripción PAX3/fisiología
8.
Nat Commun ; 10(1): 2487, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31171776

RESUMEN

Lack or excess expression of the surface ectoderm-expressed transcription factor Grainyhead-like2 (Grhl2), each prevent spinal neural tube closure. Here we investigate the causative mechanisms and find reciprocal dysregulation of epithelial genes, cell junction components and actomyosin properties in Grhl2 null and over-expressing embryos. Grhl2 null surface ectoderm shows a shift from epithelial to neuroepithelial identity (with ectopic expression of N-cadherin and Sox2), actomyosin disorganisation, cell shape changes and diminished resistance to neural fold recoil upon ablation of the closure point. In contrast, excessive abundance of Grhl2 generates a super-epithelial surface ectoderm, in which up-regulation of cell-cell junction proteins is associated with an actomyosin-dependent increase in local mechanical stress. This is compatible with apposition of the neural folds but not with progression of closure, unless myosin activity is inhibited. Overall, our findings suggest that Grhl2 plays a crucial role in regulating biomechanical properties of the surface ectoderm that are essential for spinal neurulation.


Asunto(s)
Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Tubo Neural/embriología , Células Neuroepiteliales/metabolismo , Neurulación/genética , Factores de Transcripción/genética , Actomiosina/genética , Actomiosina/metabolismo , Animales , Fenómenos Biomecánicos , Cadherinas/metabolismo , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Células Epiteliales/metabolismo , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Ratones , Tubo Neural/metabolismo , Factores de Transcripción SOXB1/metabolismo , Estrés Mecánico , Factores de Transcripción/metabolismo
9.
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
10.
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
11.
Biochimie ; 126: 63-70, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26924399

RESUMEN

The curly tail mouse provides a model for neural tube defects (spina bifida and exencephaly) that are resistant to prevention by folic acid. The major ct gene, responsible for spina bifida, corresponds to a hypomorphic allele of grainyhead-like 3 (Grhl3) but the frequency of NTDs is strongly influenced by modifiers in the genetic background. Moreover, exencephaly in the curly tail strain is not prevented by reinstatement of Grhl3 expression. In the current study we found that expression of Mthfd1L, encoding a key component of mitochondrial folate one-carbon metabolism (FOCM), is significantly reduced in ct/ct embryos compared to a partially congenic wild-type strain. This expression change is not attributable to regulation by Grhl3 or the genetic background at the Mthfd1L locus. Mitochondrial FOCM provides one-carbon units as formate for FOCM reactions in the cytosol. We found that maternal supplementation with formate prevented NTDs in curly tail embryos and also resulted in increased litter size. Analysis of the folate profile of neurulation-stage embryos showed that formate supplementation resulted in an increased proportion of formyl-THF and THF but a reduction in proportion of 5-methyl THF. In contrast, THF decreased and 5-methyl THF was relatively more abundant in the liver of supplemented dams than in controls. In embryos cultured through the period of spinal neurulation, incorporation of labelled thymidine and adenine into genomic DNA was suppressed by supplemental formate, suggesting that de novo folate-dependent biosynthesis of nucleotides (thymidylate and purines) was enhanced. We hypothesise that reduced Mthfd1L expression may contribute to susceptibility to NTDs in the curly tail strain and that formate acts as a one-carbon donor to prevent NTDs.


Asunto(s)
Ácido Fólico/metabolismo , Formiatos/farmacología , Nucleótidos/biosíntesis , Disrafia Espinal , Animales , Modelos Animales de Enfermedad , Ratones , Disrafia Espinal/metabolismo , Disrafia Espinal/prevención & control
12.
Magn Reson Med ; 73(2): 731-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24634098

RESUMEN

PURPOSE: Advanced methodologies for visualizing novel tissue contrast are essential for phenotyping the ever-increasing number of mutant mouse embryos being generated. Although diffusion microscopic MRI (µMRI) has been used to phenotype embryos, widespread routine use is limited by extended scanning times, and there is no established experimental procedure ensuring optimal data acquisition. METHODS: We developed two protocols for designing experimental procedures for diffusion µMRI of mouse embryos, which take into account the effect of embryo preparation and pulse sequence parameters on resulting data. We applied our protocols to an investigation of the splotch mouse model as an example implementation. RESULTS: The protocols provide DTI data in 24 min per direction at 75 µm isotropic using a three-dimensional fast spin-echo sequence, enabling preliminary imaging in 3 h (6 directions plus one unweighted measurement), or detailed imaging in 9 h (42 directions plus six unweighted measurements). Application to the splotch model enabled assessment of spinal cord pathology. CONCLUSION: We present guidelines for designing diffusion µMRI experiments, which may be adapted for different studies and research facilities. As they are suitable for routine use and may be readily implemented, we hope they will be adopted by the phenotyping community.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/métodos , Embrión de Mamíferos/citología , Imagen por Resonancia Magnética/métodos , Microscopía/métodos , Médula Espinal/citología , Médula Espinal/embriología , Animales , Aumento de la Imagen/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Diagnóstico Prenatal/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Manejo de Especímenes/métodos
13.
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
14.
Birth Defects Res A Clin Mol Teratol ; 97(6): 398-402, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23733478

RESUMEN

BACKGROUND: Lamins are intermediate filament proteins that form a major component of the nuclear lamina, a protein complex at the surface of the inner nuclear membrane. Numerous clinically diverse conditions, termed laminopathies, have been found to result from mutation of LMNA. In contrast, coding or loss of function mutations of LMNB1, encoding lamin B1, have not been identified in human disease. In mice, polymorphism in Lmnb1 has been shown to modify risk of neural tube defects (NTDs), malformations of the central nervous system that result from incomplete closure of the neural folds. METHODS: Mutation analysis by DNA sequencing was performed on all exons of LMNB1 in 239 samples from patients with NTDs from the United Kingdom, Sweden, and United States. Possible functional effects of missense variants were analyzed by bioinformatics prediction and fluorescence in photobleaching. RESULTS: In NTD patients, we identified two unique missense variants that were predicted to disrupt protein structure/function and represent putative contributory mutations. Fluorescence loss in photobleaching analysis showed that the A436T variant compromised stability of lamin B1 interaction within the lamina. CONCLUSION: The genetic basis of human NTDs appears highly heterogenous with possible involvement of multiple predisposing genes. We hypothesize that rare variants of LMNB1 may contribute to susceptibility to NTDs.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Lamina Tipo B/genética , Defectos del Tubo Neural/genética , Estudios de Cohortes , Biología Computacional , Análisis Mutacional de ADN , Exones/genética , Fluorescencia , Humanos , Lamina Tipo B/metabolismo , Mutación Missense/genética , Lámina Nuclear/metabolismo , Fotoblanqueo , Suecia , Reino Unido , Estados Unidos
15.
Mol Cell Biochem ; 378(1-2): 229-36, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23483428

RESUMEN

Folates act as co-factors for transfer of one-carbon units for nucleotide production, methylation and other biosynthetic reactions. Comprehensive profiling of multiple folates can be achieved using liquid chromatography tandem mass spectrometry, enabling determination of their relative abundance that may provide an indication of metabolic differences between cell types. For example, cell lines exposed to methotrexate showed a dose-dependent elevation of dihydrofolate, consistent with inhibition of dihydrofolate reductase. We analysed the folate profile of E. coli sub-types as well as cell lines and embryonic tissue from both human and mouse. The folate profile of bacteria differed markedly from those of all the mammalian samples, most notably in the greater abundance of formyl tetrahydrofolate. The overall profiles of mouse and human fibroblasts and mid-gestation mouse embryos were broadly similar, with specific differences. The major folate species in these cell types was 5-methyl tetrahydrofolate, in contrast to lymphoblastoid cell lines in which the predominant form was tetrahydrofolate. Analysis of embryonic human brain revealed a shift in folate profile with increasing developmental stage, with a decline in relative abundance of dihydrofolate and increase in 5-methyl tetrahydrofolate. These cell type-specific and developmental changes in folate profile may indicate differential requirements for the various outputs of folate metabolism.


Asunto(s)
Encéfalo/metabolismo , Tetrahidrofolatos/metabolismo , Animales , Encéfalo/embriología , Línea Celular , Escherichia coli/metabolismo , Ácido Fólico/metabolismo , Humanos , Metotrexato/química , Metotrexato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Estándares de Referencia , Especificidad de la Especie , Espectrometría de Masas en Tándem/métodos , Espectrometría de Masas en Tándem/normas , Tetrahidrofolatos/química
16.
PLoS Genet ; 8(11): e1003059, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166514

RESUMEN

Neural tube defects (NTDs), including spina bifida and anencephaly, are common birth defects whose complex multigenic causation has hampered efforts to delineate their molecular basis. The effect of putative modifier genes in determining NTD susceptibility may be investigated in mouse models, particularly those that display partial penetrance such as curly tail, a strain in which NTDs result from a hypomorphic allele of the grainyhead-like-3 gene. Through proteomic analysis, we found that the curly tail genetic background harbours a polymorphic variant of lamin B1, lacking one of a series of nine glutamic acid residues. Lamins are intermediate filament proteins of the nuclear lamina with multiple functions that influence nuclear structure, cell cycle properties, and transcriptional regulation. Fluorescence loss in photobleaching showed that the variant lamin B1 exhibited reduced stability in the nuclear lamina. Genetic analysis demonstrated that the variant also affects neural tube closure: the frequency of spina bifida and anencephaly was reduced three-fold when wild-type lamin B1 was bred into the curly tail strain background. Cultured fibroblasts expressing variant lamin B1 show significantly increased nuclear dysmorphology and diminished proliferative capacity, as well as premature senescence, associated with reduced expression of cyclins and Smc2, and increased expression of p16. The cellular basis of spinal NTDs in curly tail embryos involves a proliferation defect localised to the hindgut epithelium, and S-phase progression was diminished in the hindgut of embryos expressing variant lamin B1. These observations indicate a mechanistic link between altered lamin B1 function, exacerbation of the Grhl3-mediated cell proliferation defect, and enhanced susceptibility to NTDs. We conclude that lamin B1 is a modifier gene of major effect for NTDs resulting from loss of Grhl3 function, a role that is likely mediated via the key function of lamin B1 in maintaining integrity of the nuclear envelope and ensuring normal cell cycle progression.


Asunto(s)
Ciclo Celular , Lamina Tipo B , Defectos del Tubo Neural , Membrana Nuclear , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , División Celular , Proliferación Celular , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Ratones , Mutación , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Polimorfismo Genético , Proteómica , Disrafia Espinal/genética , Disrafia Espinal/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
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
18.
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
19.
Am J Hum Genet ; 84(2): 197-209, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19200523

RESUMEN

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous inherited disorder arising from dysmotility of motile cilia and sperm. This is associated with a variety of ultrastructural defects of the cilia and sperm axoneme that affect movement, leading to clinical consequences on respiratory-tract mucociliary clearance and lung function, fertility, and left-right body-axis determination. We performed whole-genome SNP-based linkage analysis in seven consanguineous families with PCD and central-microtubular-pair abnormalities. This identified two loci, in two families with intermittent absence of the central-pair structure (chromosome 6p21.1, Zmax 6.7) and in five families with complete absence of the central pair (chromosome 6q22.1, Zmax 7.0). Mutations were subsequently identified in two positional candidate genes, RSPH9 on chromosome 6p21.1 and RSPH4A on chromosome 6q22.1. Haplotype analysis identified a common ancestral founder effect RSPH4A mutation present in UK-Pakistani pedigrees. Both RSPH9 and RSPH4A encode protein components of the axonemal radial spoke head. In situ hybridization of murine Rsph9 shows gene expression restricted to regions containing motile cilia. Investigation of the effect of knockdown or mutations of RSPH9 orthologs in zebrafish and Chlamydomonas indicate that radial spoke head proteins are important in maintaining normal movement in motile, "9+2"-structure cilia and flagella. This effect is rescued by reintroduction of gene expression for restoration of a normal beat pattern in zebrafish. Disturbance in function of these genes was not associated with defects in left-right axis determination in humans or zebrafish.


Asunto(s)
Cilios/patología , Anomalías Congénitas/genética , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Síndrome de Kartagener/genética , Mutación , Animales , Chlamydomonas/genética , Aberraciones Cromosómicas , Mapeo Cromosómico , Cromosomas Humanos/genética , Cromosomas Humanos Par 1 , Cilios/genética , Femenino , Humanos , Hibridación in Situ , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Pez Cebra/genética
20.
Hum Mol Genet ; 17(14): 2150-9, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18407919

RESUMEN

Homozygous loss-of-function mutations in the transcription factor LHX3 have been associated with hypopituitarism with structural anterior pituitary defects and cervical abnormalities with or without restricted neck rotation. We report two novel recessive mutations in LHX3 in four patients from two unrelated pedigrees. Clinical evaluation revealed that all four patients exhibit varying degrees of bilateral sensorineural hearing loss, which has not been previously reported in association with LHX3 mutations, in addition to hypopituitarism including adrenocorticotropic hormone deficiency and an unusual skin and skeletal phenotype in one family. Furthermore, re-evaluation of three patients previously described with LHX3 mutations showed they also exhibit varying degrees of bilateral sensorineural hearing loss. We have investigated a possible role for LHX3 in inner ear development in humans using in situ hybridization of human embryonic and fetal tissue. LHX3 is expressed in defined regions of the sensory epithelium of the developing inner ear in a pattern overlapping that of SOX2, which precedes the onset of LHX3 expression and is known to be required for inner ear and pituitary development in both mice and humans. Moreover, we show that SOX2 is capable of binding to and activating transcription of the LHX3 proximal promoter in vitro. This study therefore extends the phenotypic spectrum associated with LHX3 mutations to encompass variable sensorineural hearing loss and suggests a possible interaction between LHX3 and SOX2 likely to be important for development of both the inner ear and the anterior pituitary in human embryonic development.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas de Homeodominio/genética , Hipopituitarismo/genética , Mutación , Adolescente , Animales , Secuencia de Bases , Células CHO , Niño , Cricetinae , Cricetulus , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Oído Interno/embriología , Oído Interno/crecimiento & desarrollo , Oído Interno/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Expresión Génica , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Pérdida Auditiva Sensorineural/embriología , Pérdida Auditiva Sensorineural/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Hipopituitarismo/embriología , Hipopituitarismo/metabolismo , Lactante , Proteínas con Homeodominio LIM , Masculino , Ratones , Polimorfismo de Nucleótido Simple , Factores de Transcripción SOXB1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...