Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Cardiovasc Res ; 88(2): 287-95, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20558441

ABSTRACT

AIMS: Cardiac malformations are prevalent in trisomies of human chromosome 21 [Down's syndrome (DS)], affecting normal chamber separation in the developing heart. Efforts to understand the aetiology of these defects have been severely hampered by the absence of an accurate mouse model. Such models have proved challenging to establish because synteny with human chromosome Hsa21 is distributed across three mouse chromosomes. None of those engineered so far accurately models the full range of DS cardiac phenotypes, in particular the profound disruptions resulting from atrioventricular septal defects (AVSDs). Here, we present analysis of the cardiac malformations exhibited by embryos of the transchromosomic mouse line Tc(Hsa21)1TybEmcf (Tc1) which contains more than 90% of chromosome Hsa21 in addition to the normal diploid mouse genome. METHODS AND RESULTS: Using high-resolution episcopic microscopy and three-dimensional (3D) modelling, we show that Tc1 embryos exhibit many of the cardiac defects found in DS, including balanced AVSD with single and separate valvar orifices, membranous and muscular ventricular septal defects along with outflow tract and valve leaflet abnormalities. Frequencies of cardiac malformations (ranging from 38 to 55%) are dependent on strain background. In contrast, no comparable cardiac defects were detected in embryos of the more limited mouse trisomy model, Dp(16Cbr1-ORF9)1Rhr (Ts1Rhr), indicating that trisomy of the region syntenic to the Down's syndrome critical region, including the candidate genes DSCAM and DYRK1A, is insufficient to yield DS cardiac abnormalities. CONCLUSION: The Tc1 mouse line provides a suitable model for studying the underlying genetic causes of the DS AVSD cardiac phenotype.


Subject(s)
Abnormalities, Multiple , Chromosomes, Human, Pair 21 , Down Syndrome/genetics , Endocardial Cushion Defects/genetics , Fetal Heart/abnormalities , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Ventricular/genetics , Animals , Aorta/abnormalities , Disease Models, Animal , Down Syndrome/embryology , Embryo, Mammalian/abnormalities , Endocardial Cushion Defects/embryology , Genotype , Gestational Age , Heart Septal Defects, Atrial/embryology , Heart Septal Defects, Ventricular/embryology , Humans , Imaging, Three-Dimensional , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Microscopy/methods , Morphogenesis , Phenotype
3.
Brain ; 130(Pt 4): 1043-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17438019

ABSTRACT

Folic acid supplementation can prevent many cases of neural tube defects (NTDs), whereas suboptimal maternal folate status is a risk factor, suggesting that folate metabolism is a key determinant of susceptibility to NTDs. Despite extensive genetic analysis of folate cycle enzymes, and quantification of metabolites in maternal blood, neither the protective mechanism nor the relationship between maternal folate status and susceptibility are understood in most cases. In order to investigate potential abnormalities in folate metabolism in the embryo itself, we derived primary fibroblastic cell lines from foetuses affected by NTDs and subjected them to the dU suppression test, a sensitive metabolic test of folate metabolism. Significantly, a subset of NTD cases exhibited low scores in this test, indicative of abnormalities in folate cycling that may be causally linked to the defect. Susceptibility to NTDs may be increased by suppression of the methylation cycle, which is interlinked with the folate cycle. However, reduced efficacy in the dU suppression test was not associated with altered abundance of the methylation cycle intermediates, s-adenosylmethionine and s-adenosylhomocysteine, suggesting that a methylation cycle defect is unlikely to be responsible for the observed abnormality of folate metabolism. Genotyping of samples for known polymorphisms in genes encoding folate-associated enzymes did not reveal any correlation between specific genotypes and the observed abnormalities in folate metabolism. These data suggest that as yet unrecognized genetic variants result in embryonic abnormalities of folate cycling that may be causally related to NTDs.


Subject(s)
Fetal Diseases/metabolism , Fetus/metabolism , Folic Acid/metabolism , Neural Tube Defects/metabolism , Anencephaly/embryology , Anencephaly/metabolism , Animals , Antimetabolites/pharmacology , Deoxyuridine/pharmacology , Female , Ferredoxin-NADP Reductase/genetics , Fetus/drug effects , Fibroblasts/metabolism , Folic Acid/genetics , Genotype , Humans , Methylation , Mice , NIH 3T3 Cells , Neural Tube Defects/embryology , Neural Tube Defects/genetics , Polymorphism, Genetic/genetics , Pregnancy , S-Adenosylhomocysteine/analysis , S-Adenosylmethionine/analysis , Spinal Dysraphism/embryology , Spinal Dysraphism/metabolism
4.
Birth Defects Res A Clin Mol Teratol ; 76(7): 544-52, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16933307

ABSTRACT

BACKGROUND: Closure of the cranial neural tube during embryogenesis is a crucial process in development of the brain. Failure of this event results in the severe neural tube defect (NTD) exencephaly, the developmental forerunner of anencephaly. METHODS: The requirement for methylation cycle function in cranial neural tube closure was tested by treatment of cultured mouse embryos with cycloleucine or ethionine, inhibitors of methionine adenosyl transferase. Embryonic phenotypes were investigated by histological analysis, and immunostaining was performed for markers of proliferation and apoptosis. Methylation cycle intermediates s-adenosylmethionine and s-adenosylhomocysteine were also quantitated by tandem mass spectrometry. RESULTS: Ethionine and cycloleucine treatments significantly reduced the ratio of abundance of s-adenosylmethionine to s-adenosylhomocysteine and are, therefore, predicted to suppress the methylation cycle. Exposure to these inhibitors during the period of cranial neurulation caused a high incidence of exencephaly, in the absence of generalized toxicity, growth retardation, or developmental delay. Reduced neuroepithelial thickness and reduced density of cranial mesenchyme were detected in ethionine-treated but not cycloleucine-treated embryos that developed exencephaly. Reduced mesenchymal density is a potential cause of ethionine-induced exencephaly, although we could not detect a causative alteration in proliferation or apoptosis prior to failure of neural tube closure. CONCLUSIONS: Adequate functioning of the methylation cycle is essential for cranial neural tube closure in the mouse, suggesting that suppression of the methylation cycle could also increase the risk of human NTDs. We hypothesize that inhibition of the methylation cycle causes NTDs due to disruption of crucial reactions involving methylation of DNA, proteins or other biomolecules.


Subject(s)
Neural Tube Defects/embryology , Neural Tube Defects/metabolism , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Animals , Cycloleucine/pharmacology , Embryo Culture Techniques , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Ethionine/pharmacology , Female , Male , Methylation/drug effects , Mice , Mice, Inbred Strains , Neural Tube Defects/chemically induced
5.
FEBS Lett ; 580(11): 2803-7, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16674949

ABSTRACT

Suppression of one-carbon metabolism or insufficient methionine intake are suggested to increase risk of neural tube defects (NTD). Here, exogenous methionine unexpectedly caused frequent NTD in cultured mouse embryos. NTD were associated with reduced cranial mesenchyme cell density, which may result from a preceding reduction in proliferation. The abundance ratio of S-adenosylmethionine to S-adenosylhomocysteine was also decreased in treated embryos, suggesting methylation reactions may be suppressed. Such an effect is potentially causative as NTD were also observed when DNA methylation was specifically inhibited. Thus, reduced cranial mesenchyme density and impairment of critical methylation reactions may contribute to development of methionine-induced NTD.


Subject(s)
Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Methionine/pharmacology , Neural Tube Defects/chemically induced , Neural Tube Defects/embryology , Animals , Azacitidine/pharmacology , Embryo Culture Techniques , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Folic Acid/pharmacology , Methylation/drug effects , Mice , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Phenotype
6.
Hum Mol Genet ; 13(1): 7-14, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14613966

ABSTRACT

A proportion of neural tube defects (NTDs) can be prevented by maternal folic acid supplementation, although some cases are unresponsive. The curly tail mutant mouse provides a model of folate-resistant NTDs, in which defects can be prevented by inositol therapy in early pregnancy. Hence, inositol represents a possible novel adjunct therapy to prevent human NTDs. The present study investigated the molecular mechanism by which inositol prevents mouse NTDs. Activation of protein kinase C (PKC) is known to be essential, and we examined neurulation-stage embryos for PKC expression and applied PKC inhibitors to curly tail embryos developing in culture. Although all known PKC isoforms were detected in the closing neural tube, use of chemical PKC inhibitors identified a particular requirement for 'conventional' PKC isoforms. Peptide inhibitors offer selective inhibition of individual PKCs, and we demonstrated isoform-specific inhibition of PKC in embryonic cell cultures. Application of peptide inhibitors to neurulation-stage embryos revealed an absolute dependence on the activity of PKCbetaI and gamma for prevention of NTDs by inositol, and partial dependence on PKCzeta, whereas other PKCs (alpha, betaII delta, and epsilon) were dispensable. To investigate the cellular action of inositol and PKCs in NTD prevention, we examined cell proliferation in curly tail embryos. Defective proliferation of hindgut cells is a key component of the pathogenic sequence leading to NTDs in curly tail. Hindgut cell proliferation was stimulated specifically by inositol, an effect that required activation of PKCbetaI. Our findings reveal an essential role of specific PKC isoforms in mediating the prevention of mouse NTDs by inositol.


Subject(s)
Gene Expression Regulation, Enzymologic , Inositol/therapeutic use , Neural Tube Defects/drug therapy , Protein Kinase C/metabolism , Animals , Blotting, Western , Disease Models, Animal , Embryo, Mammalian/metabolism , Immunohistochemistry , Inositol/metabolism , Isoenzymes/metabolism , Mice , Mice, Mutant Strains , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Protein Kinase C/antagonists & inhibitors
7.
Anat Embryol (Berl) ; 206(3): 185-91, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12592569

ABSTRACT

The observation of elevated maternal plasma homocysteine concentration in pregnancies affected by neural tube defects (NTD) suggests that folate metabolism may be disturbed in NTD cases. In addition, studies on the chick embryo suggest that hyperhomocysteinaemia may contribute directly to the development of NTD. In order to test the hypothesis that homocysteine may cause NTD, we cultured mouse embryos in the presence of homocysteine thiolactone during the period of cranial neural tube closure. At doses of 0.5 mM or above, exposure to homocysteine thiolactone caused growth retardation, blisters and abnormalities of somite development. Despite the teratogenic effects of homocysteine we did not detect any increase in the incidence of neural tube defects. Neither was there an effect on the incorporation of thymidine into DNA, a potential marker of alterations in the folate or homocysteine/methionine cycles. These observations suggest that homocysteine is unlikely to be a direct cause of NTD in humans. Rather, the elevated levels of homocysteine in human NTD pregnancies may reflect a disturbance in folate-related metabolism.


Subject(s)
Central Nervous System/abnormalities , Central Nervous System/drug effects , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Homocysteine/toxicity , Neural Tube Defects/chemically induced , Animals , Central Nervous System/pathology , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian/pathology , Female , Fetus , Folic Acid/metabolism , Homocysteine/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Pregnancy/metabolism , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , Somites/pathology , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...