Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
FASEB J ; 38(11): e23738, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38855924

ABSTRACT

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.


Subject(s)
Inositol , Neural Tube Defects , Inositol/metabolism , Inositol/pharmacology , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Animals , Female , Mice , Pregnancy , Embryo, Mammalian/metabolism , Maternal Nutritional Physiological Phenomena , Metabolome , Folic Acid/metabolism
2.
Mol Genet Metab ; 142(3): 108496, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761651

ABSTRACT

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.


Subject(s)
Brain , Dependovirus , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Glycine Dehydrogenase (Decarboxylating) , Glycine , Hyperglycinemia, Nonketotic , Liver , Animals , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/therapy , Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine Dehydrogenase (Decarboxylating)/metabolism , Dependovirus/genetics , Mice , Humans , Genetic Vectors/genetics , Glycine/metabolism , Liver/metabolism , Brain/metabolism , Biomarkers/metabolism , Folic Acid/metabolism
3.
Mol Cell Proteomics ; 23(3): 100718, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38224738

ABSTRACT

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.


Subject(s)
RNA , Tetrahydrofolate Dehydrogenase , Humans , Cell Line , Peptides/metabolism , Protein Biosynthesis , Ribosomes/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
4.
Hum Mol Genet ; 32(17): 2681-2692, 2023 08 26.
Article in English | MEDLINE | ID: mdl-37364051

ABSTRACT

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.


Subject(s)
Abnormalities, Multiple , Cleft Lip , Cleft Palate , Neural Tube Defects , Spinal Dysraphism , Animals , Humans , Mice , Abnormalities, Multiple/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Encephalocele/genetics , Mutation , Neural Tube Defects/genetics , Spinal Dysraphism/genetics
5.
Front Genet ; 12: 625120, 2021.
Article in English | MEDLINE | ID: mdl-33569080

ABSTRACT

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.

6.
J Inherit Metab Dis ; 43(6): 1186-1198, 2020 11.
Article in English | MEDLINE | ID: mdl-32743799

ABSTRACT

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.


Subject(s)
Brain/metabolism , Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/enzymology , Alleles , Animals , Brain/pathology , Hyperglycinemia, Nonketotic/pathology , Mice , Mutation, Missense
7.
Birth Defects Res ; 112(2): 205-211, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31758757

ABSTRACT

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.


Subject(s)
Neural Tube Defects/embryology , Neurulation/genetics , Acetylation , Anencephaly/etiology , Anencephaly/physiopathology , Animals , Disease Models, Animal , Embryonic Development/genetics , Embryonic Development/physiology , Histone Acetyltransferases/metabolism , Humans , Mammals , Mice/embryology , Neurulation/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spinal Dysraphism/etiology , Spinal Dysraphism/physiopathology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Dis Model Mech ; 12(11)2019 11 22.
Article in English | MEDLINE | ID: mdl-31636139

ABSTRACT

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.


Subject(s)
Folic Acid/administration & dosage , Mutation , Neural Tube Defects/etiology , PAX3 Transcription Factor/genetics , Animals , Apoptosis , Cell Cycle/drug effects , Dietary Supplements , Disease Models, Animal , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Neural Tube Defects/prevention & control , PAX3 Transcription Factor/physiology
9.
Nat Commun ; 10(1): 2487, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171776

ABSTRACT

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.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Neural Tube/embryology , Neuroepithelial Cells/metabolism , Neurulation/genetics , Transcription Factors/genetics , Actomyosin/genetics , Actomyosin/metabolism , Animals , Biomechanical Phenomena , Cadherins/metabolism , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/metabolism , Epithelial Cells/metabolism , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Mice , Neural Tube/metabolism , SOXB1 Transcription Factors/metabolism , Stress, Mechanical , Transcription Factors/metabolism
10.
Hum Mol Genet ; 27(24): 4218-4230, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30189017

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , Nerve Tissue Proteins/genetics , Neural Tube Defects/genetics , Spinal Dysraphism/genetics , Transcription Factors/genetics , Alleles , Animals , Animals, Genetically Modified/genetics , Disease Models, Animal , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Loss of Function Mutation , Mice , Neural Tube Defects/pathology , Protein Multimerization/genetics , Spinal Dysraphism/pathology
11.
Dev Biol ; 435(2): 130-137, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29397878

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/physiology , Neural Tube Defects/genetics , Neural Tube/physiology , Neurulation/genetics , Transcription Factors/physiology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Genes, Reporter , Germ Layers/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Plate/metabolism , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Organ Specificity , RNA, Messenger/biosynthesis , Spinal Dysraphism/embryology , Spinal Dysraphism/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
12.
Biochimie ; 126: 63-70, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26924399

ABSTRACT

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.


Subject(s)
Folic Acid/metabolism , Formates/pharmacology , Nucleotides/biosynthesis , Spinal Dysraphism , Animals , Disease Models, Animal , Mice , Spinal Dysraphism/metabolism , Spinal Dysraphism/prevention & control
13.
Magn Reson Med ; 73(2): 731-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24634098

ABSTRACT

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.


Subject(s)
Diffusion Magnetic Resonance Imaging/methods , Embryo, Mammalian/cytology , Magnetic Resonance Imaging/methods , Microscopy/methods , Spinal Cord/cytology , Spinal Cord/embryology , Animals , Image Enhancement/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Prenatal Diagnosis/methods , Reproducibility of Results , Sensitivity and Specificity , Specimen Handling/methods
14.
Brain ; 136(Pt 9): 2836-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23935126

ABSTRACT

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.


Subject(s)
Folic Acid/adverse effects , Neural Tube Defects/prevention & control , Purine Nucleosides/therapeutic use , Pyrimidine Nucleosides/therapeutic use , Animals , Body Patterning/drug effects , Body Patterning/physiology , Cell Proliferation/drug effects , Disease Models, Animal , Embryo, Mammalian , Female , Folic Acid/metabolism , Histones/metabolism , Litter Size/drug effects , Male , Maternal Exposure , Mice , Mice, Mutant Strains , Neural Tube Defects/drug therapy , Neural Tube Defects/genetics , Pregnancy , Statistics, Nonparametric , Thymidine/therapeutic use
15.
Birth Defects Res A Clin Mol Teratol ; 97(6): 398-402, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23733478

ABSTRACT

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.


Subject(s)
Genetic Predisposition to Disease/genetics , Lamin Type B/genetics , Neural Tube Defects/genetics , Cohort Studies , Computational Biology , DNA Mutational Analysis , Exons/genetics , Fluorescence , Humans , Lamin Type B/metabolism , Mutation, Missense/genetics , Nuclear Lamina/metabolism , Photobleaching , Sweden , United Kingdom , United States
16.
Mol Cell Biochem ; 378(1-2): 229-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23483428

ABSTRACT

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.


Subject(s)
Brain/metabolism , Tetrahydrofolates/metabolism , Animals , Brain/embryology , Cell Line , Escherichia coli/metabolism , Folic Acid/metabolism , Humans , Methotrexate/chemistry , Methotrexate/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Reference Standards , Species Specificity , Tandem Mass Spectrometry/methods , Tandem Mass Spectrometry/standards , Tetrahydrofolates/chemistry
17.
PLoS Genet ; 8(11): e1003059, 2012.
Article in English | MEDLINE | ID: mdl-23166514

ABSTRACT

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.


Subject(s)
Cell Cycle , Lamin Type B , Neural Tube Defects , Nuclear Envelope , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Cell Division , Cell Proliferation , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Mutation , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Polymorphism, Genetic , Proteomics , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Hum Mol Genet ; 20(8): 1536-46, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21262862

ABSTRACT

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.


Subject(s)
Spinal Dysraphism/genetics , Transcription Factors/genetics , Animals , Cell Proliferation , Chromosome Mapping , Chromosomes, Mammalian/genetics , Female , Gene Silencing , Genetic Linkage , Humans , Hybridization, Genetic , Lower Gastrointestinal Tract/abnormalities , Lower Gastrointestinal Tract/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Spinal Dysraphism/embryology , Transcription Factors/metabolism , Transcription, Genetic , Up-Regulation
19.
Birth Defects Res A Clin Mol Teratol ; 88(8): 612-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20589880

ABSTRACT

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.


Subject(s)
Carbon/metabolism , Folic Acid Deficiency/complications , Folic Acid/metabolism , Neural Tube Defects/etiology , Animals , DNA-Binding Proteins/genetics , Female , Fibroblasts/metabolism , Folic Acid Deficiency/genetics , Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice , Mice, Mutant Strains , Neural Tube Defects/metabolism , Pregnancy , S-Adenosylhomocysteine/analysis , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/analysis , S-Adenosylmethionine/metabolism , Thymidine Monophosphate/biosynthesis , Transcription Factors/genetics
20.
Am J Hum Genet ; 84(2): 197-209, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19200523

ABSTRACT

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.


Subject(s)
Cilia/pathology , Congenital Abnormalities/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Kartagener Syndrome/genetics , Mutation , Animals , Chlamydomonas/genetics , Chromosome Aberrations , Chromosome Mapping , Chromosomes, Human/genetics , Chromosomes, Human, Pair 1 , Cilia/genetics , Female , Humans , In Situ Hybridization , Male , Pedigree , Polymorphism, Single Nucleotide , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL