Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 9737, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37322040

ABSTRACT

Dominantly inherited GAA repeat expansions in FGF14 are a common cause of spinocerebellar ataxia (GAA-FGF14 ataxia; spinocerebellar ataxia 27B). Molecular confirmation of FGF14 GAA repeat expansions has thus far mostly relied on long-read sequencing, a technology that is not yet widely available in clinical laboratories. We developed and validated a strategy to detect FGF14 GAA repeat expansions using long-range PCR, bidirectional repeat-primed PCRs, and Sanger sequencing. We compared this strategy to targeted nanopore sequencing in a cohort of 22 French Canadian patients and next validated it in a cohort of 53 French index patients with unsolved ataxia. Method comparison showed that capillary electrophoresis of long-range PCR amplification products significantly underestimated expansion sizes compared to nanopore sequencing (slope, 0.87 [95% CI, 0.81 to 0.93]; intercept, 14.58 [95% CI, - 2.48 to 31.12]) and gel electrophoresis (slope, 0.84 [95% CI, 0.78 to 0.97]; intercept, 21.34 [95% CI, - 27.66 to 40.22]). The latter techniques yielded similar size estimates. Following calibration with internal controls, expansion size estimates were similar between capillary electrophoresis and nanopore sequencing (slope: 0.98 [95% CI, 0.92 to 1.04]; intercept: 10.62 [95% CI, - 7.49 to 27.71]), and gel electrophoresis (slope: 0.94 [95% CI, 0.88 to 1.09]; intercept: 18.81 [95% CI, - 41.93 to 39.15]). Diagnosis was accurately confirmed for all 22 French Canadian patients using this strategy. We also identified 9 French patients (9/53; 17%) and 2 of their relatives who carried an FGF14 (GAA)≥250 expansion. This novel strategy reliably detected and sized FGF14 GAA expansions, and compared favorably to long-read sequencing.


Subject(s)
Friedreich Ataxia , Spinocerebellar Ataxias , Humans , Canada , Friedreich Ataxia/genetics , Spinocerebellar Ataxias/genetics , Trinucleotide Repeat Expansion
2.
Int J Mol Sci ; 24(12)2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37372992

ABSTRACT

Vitamin B9 (folate)/B12 (cobalamin) deficiency is known to induce brain structural and/or functional retardations. In many countries, folate supplementation, targeting the most severe outcomes such as neural tube defects, is discontinued after the first trimester. However, adverse effects may occur after birth because of some mild misregulations. Various hormonal receptors were shown to be deregulated in brain tissue under these conditions. The glucocorticoid receptor (GR) is particularly sensitive to epigenetic regulation and post-translational modifications. In a mother-offspring rat model of vitamin B9/B12 deficiency, we investigated whether a prolonged folate supplementation could restore the GR signaling in the hypothalamus. Our data showed that a deficiency of folate and vitamin B12 during the in-utero and early postnatal periods was associated with reduced GR expression in the hypothalamus. We also described for the first time a novel post-translational modification of GR that impaired ligand binding and GR activation, leading to decrease expression of one of the GR targets in the hypothalamus, AgRP. Moreover, this brain-impaired GR signaling pathway was associated with behavioral perturbations during offspring growth. Importantly, perinatal and postnatal supplementation with folic acid helped restore GR mRNA levels and activity in hypothalamus cells and improved behavioral deficits.


Subject(s)
Folic Acid , Vitamin B 12 Deficiency , Pregnancy , Female , Animals , Rats , Folic Acid/pharmacology , Receptors, Glucocorticoid/genetics , Glucocorticoids , Epigenesis, Genetic , Dietary Supplements , Vitamin B 12/pharmacology , Hypothalamus
3.
Cells ; 12(9)2023 04 27.
Article in English | MEDLINE | ID: mdl-37174668

ABSTRACT

Impairment of one-carbon metabolism during pregnancy, either due to nutritional deficiencies in B9 or B12 vitamins or caused by specific genetic defects, is often associated with neurological defects, including cognitive dysfunction that persists even after vitamin supplementation. Animal nutritional models do not allow for conclusions regarding the specific brain mechanisms that may be modulated by systemic compensations. Using the Cre-lox system associated to the neuronal promoter Thy1.2, a knock-out model for the methionine synthase specifically in the brain was generated. Our results on the neurobehavioral development of offspring show that the absence of methionine synthase did not lead to growth retardation, despite an effective reduction of both its expression and the methylation status in brain tissues. Behaviors were differently affected according to their functional outcome. Only temporary retardations were recorded in the acquisition of vegetative functions during the suckling period, compared to a dramatic reduction in cognitive performance after weaning. Investigation of the glutamatergic synapses in cognitive areas showed a reduction of AMPA receptors phosphorylation and clustering, indicating an epigenomic effect of the neuronal deficiency of methionine synthase on the reduction of glutamatergic synapses excitability. Altogether, our data indicate that cognitive impairment associated with methionine synthase deficiency may not only result from neurodevelopmental abnormalities, but may also be the consequence of alterations in functional plasticity of the brain.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Cognitive Dysfunction , Mice , Pregnancy , Animals , Female , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Vitamin B 12
4.
Neurogenetics ; 23(4): 241-255, 2022 10.
Article in English | MEDLINE | ID: mdl-35788923

ABSTRACT

ATL1-related spastic paraplegia SPG3A is a pure form of hereditary spastic paraplegia. Rare complex phenotypes have been described, but few data concerning cognitive evaluation or molecular imaging of these patients are available. We relate a retrospective collection of patients with SPG3A from the Neurology Department of Nancy University Hospital, France. For each patient were carried out a 18F-FDG PET (positron emission tomography), a electromyography (EMG), a sudoscan®, a cerebral and spinal cord MRI (magnetic resonance imaging) with measurement of cervical and thoracic surfaces, a neuropsychological assessment. The present report outlines standardised clinical and paraclinical data of five patients from two east-France families carrying the same missense pathogenic variation, NM_015915.4(ATL1): c.1483C > T p.(Arg495Trp) in ATL1. Mean age at onset was 14 ± 15.01 years. Semi-quantitatively and in comparison to healthy age-matched subjects, PET scans showed a significant cerebellar and upper or mild temporal hypometabolism in all four adult patients and hypometabolism of the prefrontal cortex or precuneus in three of them. Sudoscan® showed signs of small fibre neuropathy in three patients. Cervical and thoracic patients' spinal cords were significantly thinner than matched-control, respectively 71 ± 6.59mm2 (p = 0.01) and 35.64 ± 4.35mm2 (p = 0.015). Two patients presented with a dysexecutive syndrome. While adding new clinical and paraclinical signs associated with ATL1 pathogenic variations, we insist here on the variable penetrance and expressivity. We report small fibre neuropathy, cerebellar hypometabolism and dysexecutive syndromes associated with SPG3A. These cognitive impairments and PET findings may be related to a cortico-cerebellar bundle axonopathy described in the cerebellar cognitive affective syndrome (CCAS).


Subject(s)
Small Fiber Neuropathy , Spastic Paraplegia, Hereditary , Humans , Spastic Paraplegia, Hereditary/diagnostic imaging , Spastic Paraplegia, Hereditary/genetics , Fluorodeoxyglucose F18 , DNA Mutational Analysis , Penetrance , Retrospective Studies , Pedigree , GTP-Binding Proteins/genetics , Membrane Proteins/genetics , Mutation , Phenotype , Brain/diagnostic imaging
5.
J Neurol ; 268(5): 1927-1937, 2021 May.
Article in English | MEDLINE | ID: mdl-33417001

ABSTRACT

BACKGROUND: STUB1 has been first associated with autosomal recessive (SCAR16, MIM# 615768) and later with dominant forms of ataxia (SCA48, MIM# 618093). Pathogenic variations in STUB1 are now considered a frequent cause of cerebellar ataxia. OBJECTIVE: We aimed to improve the clinical, radiological, and molecular delineation of SCAR16 and SCA48. METHODS: Retrospective collection of patients with SCAR16 or SCA48 diagnosed in three French genetic centers (Montpellier, Strasbourg and Nancy). RESULTS: Here, we report four SCAR16 and nine SCA48 patients from two SCAR16 and five SCA48 unrelated French families. All presented with slowly progressive cerebellar ataxia. Additional findings included cognitive decline, dystonia, parkinsonism and swallowing difficulties. The age at onset was highly variable, ranging from 14 to 76 years. Brain MRI showed marked cerebellar atrophy in all patients. Phenotypic findings associated with STUB1 pathogenic variations cover a broad spectrum, ranging from isolated slowly progressive ataxia to severe encephalopathy, and include extrapyramidal features. We described five new pathogenic variations, two previously reported pathogenic variations, and two rare variants of unknown significance in association with STUB1-related disorders. We also report the first pathogenic variation associated with both dominant and recessive forms of inheritance (SCAR16 and SCA48). CONCLUSION: Even though differences are observed between the recessive and dominant forms, it appears that a continuum exists between these two entities. While adding new symptoms associated with STUB1 pathogenic variations, we insist on the difficulty of genetic counselling in STUB1-related pathologies. Finally, we underscore the usefulness of DAT-scan as an additional clue for diagnosis.


Subject(s)
Cerebellar Ataxia , Ataxia , Heat-Shock Proteins , Humans , Mutation/genetics , Retrospective Studies , Ubiquitin-Protein Ligases/genetics
6.
Mol Neurobiol ; 58(3): 1024-1035, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33078371

ABSTRACT

Cobalamin (Cbl, vitamin B12) deficiency or inborn errors of Cbl metabolism can produce neurologic disorders resistant to therapies, including cognitive dysfunction, mild mental retardation, memory impairment, and confusion. We used Cd320 KO mouse as a model for studying the pathological mechanisms of these disorders. Cd320 encodes the receptor (TCblR) needed for the cellular uptake of Cbl in the brain. The Cd320-/- mouse model presented an impaired learning memory that could be alleviated by a moderate stress, which produced also a greater increase of plasma corticosterone, compared to wild type animals. The present study investigated such a putative rescue mechanism in Cbl-deficient mice. At the molecular level in the brain of Cd320-/- mouse, the decreased methylation status led to a downregulation of glucocorticoid nuclear receptor (GR)/PPAR-gamma co-activator-1 alpha (PGC-1α) pathway. This was evidenced by the decreased expression of GR, decreased methylation of GR and PGC1α, and decreased dimerization and interaction of GR with PGC1α. This led to altered synaptic activity evidenced by decreased interaction between the NMDA glutamatergic receptor and the PSD95 post-synaptic protein and a lower expression of Egr-1 and synapsin 1, in Cd320-/- mice compared to the wild type animals. Intraperitoneal injection of hydrocortisone rescued these molecular changes and normalized the learning memory tests. Our study suggests adaptive influences of moderate stress on loss of memory and cognition due to brain Cbl deficiency. The GR pathway could be a potential target for innovative therapy of cognitive manifestations in patients with poor response to conventional Cbl treatment.


Subject(s)
Brain/physiopathology , Hippocampus/physiopathology , Memory , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Vitamin B 12 Deficiency/physiopathology , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Disease Models, Animal , Glucocorticoids/pharmacology , Hippocampus/drug effects , Hydrocortisone/administration & dosage , Hydrocortisone/pharmacology , Male , Mice, Knockout , Neuronal Plasticity/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects
7.
Int J Mol Sci ; 21(19)2020 09 30.
Article in English | MEDLINE | ID: mdl-33008128

ABSTRACT

Among the numerous candidates for cell therapy of the central nervous system (CNS), olfactory progenitors (OPs) represent an interesting alternative because they are free of ethical concerns, are easy to collect, and allow autologous transplantation. In the present study, we focused on the optimization of neuron production and maturation. It is known that plated OPs respond to various trophic factors, and we also showed that the use of Nerve Growth Factor (NGF) allowed switching from a 60/40 neuron/glia ratio to an 80/20 one. Nevertheless, in order to focus on the integration of OPs in mature neural circuits, we cocultured OPs in primary cultures obtained from the cortex and hippocampus of newborn mice. When dissociated OPs were plated, they differentiated into both glial and neuronal phenotypes, but we obtained a 1.5-fold higher viability in cortex/OP cocultures than in hippocampus/OP ones. The fate of OPs in cocultures was characterized with different markers such as BrdU, Map-2, and Synapsin, indicating a healthy integration. These results suggest that the integration of transplanted OPs might by affected by trophic factors and the environmental conditions/cell phenotypes of the host tissue. Thus, a model of coculture could provide useful information on key cell events for the use of progenitors in cell therapy.


Subject(s)
Brain/metabolism , Neurons/metabolism , Olfactory Cortex/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Animals , Brain/cytology , Brain/growth & development , Cell Differentiation/genetics , Cell Lineage/genetics , Central Nervous System/metabolism , Coculture Techniques , Humans , Mice , Nerve Growth Factor/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/transplantation , Neurons/transplantation , Olfactory Cortex/cytology , Olfactory Cortex/transplantation , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/transplantation , Stem Cells/metabolism
8.
Int J Mol Sci ; 21(21)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126444

ABSTRACT

A deficiency in B-vitamins is known to lead to persistent developmental defects in various organs during early life. The nervous system is particularly affected with functional retardation in infants and young adults. In addition, even if in some cases no damage appears evident in the beginning of life, correlations have been shown between B-vitamin metabolism and neurodegenerative diseases. However, despite the usual treatment based on B-vitamin injections, the neurological outcomes remain poorly rescued in the majority of cases, compared with physiological functions. In this study, we explored whether a neonatal stimulation of neurogenesis could compensate atrophy of specific brain areas such as the hippocampus, in the case of B-vitamin deficiency. Using a physiological mild transient hypoxia within the first 24 h after birth, rat-pups, submitted or not to neonatal B-vitamin deficiency, were followed until 330-days-of-age for their cognitive capacities and their hippocampus status. Our results showed a gender effect since females were more affected than males by the deficiency, showing a persistent low body weight and poor cognitive performance to exit a maze. Nevertheless, the neonatal stimulation of neurogenesis with hypoxia rescued the maze performance during adulthood without modifying physiological markers, such as body weight and circulating homocysteine. Our findings were reinforced by an increase of several markers at 330-days-of-age in hypoxic animals, such as Ammon's Horn 1hippocampus (CA1) thickness and the expression of key actors of synaptic dynamic, such as the NMDA-receptor-1 (NMDAR1) and the post-synaptic-density-95 (PSD-95). We have not focused our conclusion on the neonatal hypoxia as a putative treatment, but we have discussed that, in the case of neurologic retardation associated with a reduced B-vitamin status, stimulation of the latent neurogenesis in infants could ameliorate their quality of life during their lifespan.


Subject(s)
Aging/pathology , Behavior, Animal , Cognitive Dysfunction/prevention & control , Folic Acid/metabolism , Neurogenesis , Vitamin B 12 Deficiency/complications , Animals , Animals, Newborn , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Female , Male , Maze Learning , Pregnancy , Rats , Rats, Wistar , Vitamin B 12/metabolism , Vitamins/metabolism
9.
Int J Mol Sci ; 20(22)2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31739389

ABSTRACT

Deficiencies in methyl donors, folate, and vitamin B12 are known to lead to brain function defects. Fetal development is the most studied but data are also available for such an impact in elderly rats. To compare the functional consequences of nutritional deficiency in young versus adult rats, we monitored behavioral outcomes of cerebellum and hippocampus circuits in the offspring of deficient mother rats and in adult rats fed a deficient diet from 2 to 8 months-of-age. We present data showing that the main deleterious consequences are found in young ages compared to adult ones, in terms of movement coordination and learning abilities. Moreover, we obtained sex and age differences in the deleterious effects on these functions and on neuronal layer integrity in growing young rats, while deficient adults presented only slight functional alterations without tissue damage. Actually, the cerebellum and the hippocampus develop and maturate according to different time lap windows and we demonstrate that a switch to a normal diet can only rescue circuits that present a long permissive window of time, such as the cerebellum, whereas the hippocampus does not. Thus, we argue, as others have, for supplements or fortifications given over a longer time than the developmental period.


Subject(s)
Brain/metabolism , Brain/physiopathology , Deficiency Diseases/complications , Deficiency Diseases/metabolism , Fetal Development , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Animals , Cognition , Deficiency Diseases/etiology , Diet , Disease Models, Animal , Female , Folic Acid Deficiency/complications , Folic Acid Deficiency/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Maze Learning , Rats
10.
Int J Mol Sci ; 20(20)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615150

ABSTRACT

The micronutrients vitamins B9 and B12 act as methyl donors in the one-carbon metabolism involved in transmethylation reactions which critically influence epigenetic mechanisms and gene expression. Both vitamins are essential for proper development, and their deficiency during pregnancy has been associated with a wide range of disorders, including persisting growth retardation. Energy homeostasis and feeding are centrally regulated by the hypothalamus which integrates peripheral signals and acts through several orexigenic and anorexigenic mediators. We studied this regulating system in a rat model of methyl donor deficiency during gestation and lactation. At weaning, a predominance of the anorexigenic pathway was observed in deficient pups, with increased plasma peptide YY and increased hypothalamic pro-opiomelanocortin (POMC) mRNA, in line with abnormal leptin, ghrelin, and insulin secretion and/or signaling during critical periods of fetal and/or postnatal development of the hypothalamus. These results suggest that early methyl donor deficiency can affect the development and function of energy balance circuits, resulting in growth and weight deficits. Maternal administration of folic acid (3 mg/kg/day) during the perinatal period tended to rectify peripheral metabolic signaling and central neuropeptide and receptor expression, leading to reduced growth retardation.


Subject(s)
Energy Metabolism/genetics , Ghrelin/genetics , Peptide YY/genetics , Pro-Opiomelanocortin/genetics , Animals , Appetite Depressants/pharmacology , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Folic Acid/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Ghrelin/blood , Hypothalamus/metabolism , Insulin/blood , Insulin/genetics , Lactation , Leptin/blood , Leptin/genetics , Methylation/drug effects , Peptide YY/blood , Pregnancy , Pro-Opiomelanocortin/blood , RNA, Messenger/genetics , Rats , Vitamin B 12/genetics , Vitamin B 12/pharmacology
11.
Int J Mol Sci ; 20(4)2019 Feb 23.
Article in English | MEDLINE | ID: mdl-30813413

ABSTRACT

Vitamins B9 (folate) and B12 act as methyl donors in the one-carbon metabolism which influences epigenetic mechanisms. We previously showed that an embryofetal deficiency of vitamins B9 and B12 in the rat increased brain expression of let-7a and miR-34a microRNAs involved in the developmental control of gene expression. This was reversed by the maternal supply with folic acid (3 mg/kg/day) during the last third of gestation, resulting in a significant reduction of associated birth defects. Since the postnatal brain is subject to intensive developmental processes, we tested whether further folate supplementation during lactation could bring additional benefits. Vitamin deficiency resulted in weaned pups (21 days) in growth retardation, delayed ossification, brain atrophy and cognitive deficits, along with unchanged brain level of let-7a and decreased expression of miR-34a and miR-23a. Whereas maternal folic acid supplementation helped restore the levels of affected microRNAs, it led to a reduction of structural and functional defects taking place during the perinatal/postnatal periods, such as learning/memory capacities. Our data suggest that a gestational B-vitamin deficiency could affect the temporal control of the microRNA regulation required for normal development. Moreover, they also point out that the continuation of folate supplementation after birth may help to ameliorate neurological symptoms commonly associated with developmental deficiencies in folate and B12.


Subject(s)
Dietary Supplements , Folic Acid/pharmacology , Growth and Development/drug effects , Animals , Behavior, Animal , Disease Models, Animal , Female , Folic Acid/blood , Homocysteine/blood , Methylation , MicroRNAs/genetics , MicroRNAs/metabolism , Nervous System/growth & development , Pregnancy , Rats, Wistar , Vitamin B 12/blood
12.
J Pathol ; 248(3): 291-303, 2019 07.
Article in English | MEDLINE | ID: mdl-30734924

ABSTRACT

The pathomechanisms that associate a deficit in folate and/or vitamin B12 and the subsequent hyperhomocysteinemia with pathological brain ageing are unclear. We investigated the homocysteinylation of microtubule-associated proteins (MAPs) in brains of patients with Alzheimer's disease or vascular dementia, and in rats depleted in folate and vitamin B12, Cd320 KO mice with selective B12 brain deficiency and H19-7 neuroprogenitors lacking folate. Compared with controls, N-homocysteinylated tau and MAP1 were increased and accumulated in protein aggregates and tangles in the cortex, hippocampus and cerebellum of patients and animals. N-homocysteinylation dissociated tau and MAPs from ß-tubulin, and MS analysis showed that it targets lysine residues critical for their binding to ß-tubulin. N-homocysteinylation increased in rats exposed to vitamin B12 and folate deficit during gestation and lactation and remained significantly higher when they became 450 days-old, despite returning to normal diet at weaning, compared with controls. It was correlated with plasma homocysteine (Hcy) and brain expression of methionine tRNAsynthetase (MARS), the enzyme required for the synthesis of Hcy-thiolactone, the substrate of N-homocysteinylation. Experimental inactivation of MARS prevented the N-homocysteinylation of tau and MAP1, and the dissociation of tau and MAP1 from ß-tubulin and PSD95 in cultured neuroprogenitors. In conclusion, increased N-homocysteinylation of tau and MAP1 is a mechanism of brain ageing that depends on Hcy concentration and expression of MARS enzyme. Its irreversibility and cumulative occurrence throughout life may explain why B12 and folate supplementation of the elderly has limited effects, if any, to prevent pathological brain ageing and cognitive decline. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Alzheimer Disease/pathology , Dementia, Vascular/pathology , Hyperhomocysteinemia/pathology , tau Proteins/metabolism , Aging/physiology , Alzheimer Disease/metabolism , Animals , Autopsy/methods , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Dementia, Vascular/metabolism , Female , Humans , Mice, Knockout , Rats
13.
Mol Neurobiol ; 54(7): 5017-5033, 2017 09.
Article in English | MEDLINE | ID: mdl-27534418

ABSTRACT

The micronutrients folate and vitamin B12 are essential for the proper development of the central nervous system, and their deficiency during pregnancy has been associated with a wide range of disorders. They act as methyl donors in the one-carbon metabolism which critically influences epigenetic mechanisms. In order to depict further underlying mechanisms, we investigated the role of let-7 and miR-34, two microRNAs regulated by methylation, on a rat model of maternal deficiency. In several countries, public health policies recommend periconceptional supplementation with folic acid. However, the question about the duration and periodicity of supplementation remains. We therefore tested maternal supply (3 mg/kg/day) during the last third of gestation from embryonic days (E) 13 to 20. Methyl donor deficiency-related developmental disorders at E20, including cerebellar and interhemispheric suture defects and atrophy of selective cerebral layers, were associated with increased brain expression (by 2.5-fold) of let-7a and miR-34a, with subsequent downregulation of their regulatory targets such as Trim71 and Notch signaling partners, respectively. These processes could be reversed by siRNA strategy in differentiating neuroprogenitors lacking folate, with improvement of their morphological characteristics. While folic acid supplementation helped restoring the levels of let-7a and miR-34a and their downstream targets, it led to a reduction of structural and functional defects taking place during the perinatal period. Our data outline the potential role of let-7 and miR-34 and their related signaling pathways in the developmental defects following gestational methyl donor deficiency and support the likely usefulness of late folate supplementation in at risk women.


Subject(s)
Brain Diseases/drug therapy , Brain/metabolism , Dietary Supplements , Folic Acid/pharmacology , MicroRNAs/metabolism , Animals , Brain Diseases/embryology , Brain Diseases/genetics , Female , MicroRNAs/drug effects , MicroRNAs/genetics , Pregnancy , Rats, Wistar
14.
FASEB J ; 29(9): 3713-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26018677

ABSTRACT

Deficiency in the methyl donors vitamin B12 and folate during pregnancy and postnatal life impairs proper brain development. We studied the consequences of this combined deficiency on cerebellum plasticity in offspring from rat mothers subjected to deficient diet during gestation and lactation and in rat neuroprogenitor cells expressing cerebellum markers. The major proteomic change in cerebellum of 21-d-old deprived females was a 2.2-fold lower expression of synapsins, which was confirmed in neuroprogenitors cultivated in the deficient condition. A pathway analysis suggested that these proteomic changes were related to estrogen receptor α (ER-α)/Src tyrosine kinase. The influence of impaired ER-α pathway was confirmed by abnormal negative geotaxis test at d 19-20 and decreased phsophorylation of synapsins in deprived females treated by ER-α antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP). This effect was consistent with 2-fold decreased expression and methylation of ER-α and subsequent decreased ER-α/PPAR-γ coactivator 1 α (PGC-1α) interaction in deficiency condition. The impaired ER-α pathway led to decreased expression of synapsins through 2-fold decreased EGR-1/Zif-268 transcription factor and to 1.7-fold reduced Src-dependent phosphorylation of synapsins. The treatment of neuroprogenitors with either MPP or PP1 (4-(4'-phenoxyanilino)-6,7-dimethoxyquinazoline, 6,7-dimethoxy-N-(4-phenoxyphenyl)-4-quinazolinamine, SKI-1, Src-l1) Src inhibitor produced similar effects. In conclusion, the deficiency during pregnancy and lactation impairs the expression of synapsins through a deregulation of ER-α pathway.


Subject(s)
Brain/metabolism , Estrogen Receptor alpha/metabolism , Folic Acid Deficiency , Gene Expression Regulation, Developmental , Lactation , Synapsins/biosynthesis , Vitamin B 12 Deficiency , Animals , Brain/embryology , Brain/pathology , Early Growth Response Protein 1/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Female , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , PPAR gamma/metabolism , Pregnancy , Rats
15.
FASEB J ; 27(6): 2185-97, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23401564

ABSTRACT

We examined the gastric mucosa structure and inflammatory status in control well-nourished Wistar dams and in Wistar dams deprived of choline, folate, and vitamin B12 during gestation and suckling periods, and in their offspring just before birth and at weaning. In this model of methyl donor deficiency (MDD), structural protein (E-cadherin and actin) N-homocysteinylation was measured through immunoprecipitation and proximity ligation assays. Cellular stress, inflammation, and apoptosis were estimated by the analysis of the NF-κB pathway, and the expression of superoxide dismutase, cyclooxygenase-2, tumor necrosis factor α, caspases 3 and 9, and TUNEL assay. Aberrant gastric mucosa formation and signs of surface layer erosion were detected in MDD fetuses and weanlings. E-cadherin and actin were N-homocysteinylated (+215 and +249% vs. controls, respectively; P<0.001). Expression of ß-catenin staining drastically decreased (-98%; P<0.01). NF-κB pathway was activated (+124%; P<0.01). Expressions of all inflammatory factors (+70%; P<0.01), superoxide dismutase (+55%; P<0.01), and caspases (+104%; P<0.01) were markedly increased. These changes were also observed in dams, to a lesser extent. Early MDD induced gastric mucosa injury similar to atrophic gastritis through structural protein N-homocysteinylation, marked inflammation, and apoptosis, despite activation of repair machinery.


Subject(s)
Cytoskeletal Proteins/metabolism , Gastritis/etiology , Gastritis/pathology , Homocysteine/metabolism , Inflammation/etiology , Inflammation/pathology , Oxidative Stress/physiology , Animals , Animals, Newborn , Animals, Suckling , Cadherins/metabolism , Female , Fetus , Gastritis/metabolism , Inflammation/metabolism , Methylation , Mothers , Pregnancy , Rats , Rats, Wistar , Severity of Illness Index , Signal Transduction/physiology
16.
Br J Nutr ; 109(4): 667-77, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-22794784

ABSTRACT

Dietary methyl donors and their genetic determinants are associated with Crohn's disease risk. We investigated whether a methyl-deficient diet (MDD) may affect development and functions of the small intestine in rat pups from dams subjected to the MDD during gestation and lactation. At 1 month before pregnancy, adult females were fed with either a standard food or a diet without vitamin B12, folate and choline. A global wall hypotrophy was observed in the distal small bowel (MDD animals 0·30 mm v. controls 0·58 mm; P< 0·001) with increased crypt apoptosis (3·37 v. 0·4%; P< 0·001), loss of enterocyte differentiation in the villus and a reduction in intestinal alkaline phosphatase production. Cleaved caspase-3 immunostaining (MDD animals 3·37% v. controls 0·4%, P< 0·001) and the Apostain labelling index showed increased crypt apoptosis (3·5 v. 1·4%; P= 0·018). Decreased proliferation was observed in crypts of the proximal small bowel with a reduced number of minichromosome maintenance 6 (MDD animals 52·83% v. controls 83·17%; P= 0·048) and proliferating cell nuclear antigen-positive cells (46·25 v. 59 %; P= 0·05). This lack of enterocyte differentiation in the distal small bowel was associated with an impaired expression of ß-catenin and a decreased ß-catenin-E-cadherin interaction. The MDD affected the intestinal barrier in the proximal small bowel by decreasing Paneth cell number after immunostaining for lysosyme (MDD animals 8·66% v. controls 21·66%) and by reducing goblet cell number and mucus production after immunostaining for mucin-2 (crypts 8·66 v. 15·33%; villus 7 v. 17%). The MDD has dual effects on the small intestine by producing dramatic effects on enterocyte differentiation and barrier function in rats.


Subject(s)
Choline Deficiency/metabolism , Enterocytes/cytology , Folic Acid Deficiency/metabolism , Intestine, Small/pathology , Vitamin B 12 Deficiency/metabolism , Alkaline Phosphatase/metabolism , Animal Feed , Animals , Apoptosis , Cadherins/metabolism , Caspase 3/metabolism , Cell Differentiation , Choline/metabolism , Female , Folic Acid/metabolism , Gene Expression Regulation , Muramidase/metabolism , Paneth Cells/metabolism , Rats , Rats, Wistar , Time Factors , Vitamin B 12/metabolism , beta Catenin/metabolism
17.
PLoS One ; 7(11): e48828, 2012.
Article in English | MEDLINE | ID: mdl-23173039

ABSTRACT

Whereas brief acute or intermittent episodes of hypoxia have been shown to exert a protective role in the central nervous system and to stimulate neurogenesis, other studies suggest that early hypoxia may constitute a risk factor that influences the future development of mental disorders. We therefore investigated the effects of a neonatal "conditioning-like" hypoxia (100% N2, 5 min) on the brain and the cognitive outcomes of rats until 720 days of age (physiologic senescence). We confirmed that such a short hypoxia led to brain neurogenesis within the ensuing weeks, along with reduced apoptosis in the hippocampus involving activation of Erk1/2 and repression of p38 and death-associated protein (DAP) kinase. At 21 days of age, increased thicknesses and cell densities were recorded in various subregions, with strong synapsin activation. During aging, previous exposure to neonatal hypoxia was associated with enhanced memory retrieval scores specifically in males, better preservation of their brain integrity than controls, reduced age-related apoptosis, larger hippocampal cell layers, and higher expression of glutamatergic and GABAergic markers. These changes were accompanied with a marked expression of synapsin proteins, mainly of their phosphorylated active forms which constitute major players of synapse function and plasticity, and with increases of their key regulators, i.e. Erk1/2, the transcription factor EGR-1/Zif-268 and Src kinase. Moreover, the significantly higher interactions between PSD-95 scaffolding protein and NMDA receptors measured in the hippocampus of 720-day-old male animals strengthen the conclusion of increased synaptic functional activity and plasticity associated with neonatal hypoxia. Thus, early non-injurious hypoxia may trigger beneficial long term effects conferring higher resistance to senescence in aged male rats, with a better preservation of cognitive functions.


Subject(s)
Aging/physiology , Brain/physiopathology , Hypoxia/physiopathology , Aging/metabolism , Animals , Animals, Newborn , Blood Gas Analysis , Brain/metabolism , Brain/pathology , Cell Death , Cell Proliferation , Cell Survival , Female , Hypoxia/blood , Hypoxia/metabolism , Hypoxia/pathology , Locomotion/physiology , Male , Memory/physiology , Neurogenesis , Neuronal Plasticity/physiology , Rats , Rats, Wistar , Synapses/metabolism
18.
FASEB J ; 26(10): 3980-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22713523

ABSTRACT

Despite the key role in neuronal development of a deficit in the methyl donor folate, little is known on the underlying mechanisms. We therefore studied the consequences of folate deficiency on proliferation, differentiation, and plasticity of the rat H19-7 hippocampal cell line. Folate deficit reduced proliferation (17%) and sensitized cells to differentiation-associated apoptosis (+16%). Decreased production (-58%) of S-adenosylmethionine (the universal substrate for transmethylation reactions) and increased expression of histone deacetylases (HDAC4,6,7) would lead to epigenomic changes that may impair the differentiation process. Cell polarity, vesicular transport, and synaptic plasticity were dramatically affected, with poor neurite outgrowth (-57%). Cell treatment by an HDAC inhibitor (SAHA) led to a noticeable improvement of cell polarity and morphology, with longer processes. Increased homocysteine levels (+55%) consecutive to folate shortage produced homocysteinylation, evidenced by coimmunoprecipitations and mass spectrometry, and aggregation of motor proteins dynein and kinesin, along with functional alterations, as reflected by reduced interactions with partner proteins. Prominent homocysteinylation of key neuronal proteins and subsequent aggregation certainly constitute major adverse effects of folate deficiency, affecting normal development with possible long-lasting consequences.


Subject(s)
Folic Acid Deficiency/metabolism , Folic Acid/pharmacology , Hippocampus/cytology , Homocysteine/pharmacology , Neurons/cytology , Neurons/drug effects , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Hep G2 Cells , Humans , Immunohistochemistry , Neurons/metabolism , Protein Binding , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Vitamin B 12/pharmacology
19.
Nutrition ; 28(9): 932-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22554959

ABSTRACT

OBJECTIVE: Diets rich in protein are often used for weight loss in obese patients, but their long-term effects are not fully understood. Homocysteine (Hcy) is considered to be a risk factor for cardiovascular diseases, and its levels are influenced by diet, particularly the protein and fat content. We studied the effect of diets with varying fat/protein content on body weight and composition, food intake, Hcy, B vitamins, leptin, and several pro-inflammatory cytokines. METHODS: For 2 mo, Long-Evans rats were fed either a low protein/high fat (LP), a standard control (C), or a high protein/low fat (HP) diet containing 5, 15, or 40% protein, respectively, and normal carbohydrate content (55% of total energy). RESULTS: The HP rats ingested 12 to 15% fewer calories (P < 0.001), gained less weight (P < 0.04), and were less fatty (P < 0.01) than the other groups. Plasma Hcy was increased in HP rats compared to C (+23%) and LP (+29%) rats (P < 0.03). It was positively correlated with protein intake (r = 0.386; P < 0.01). No obvious signs of inflammation were observed in any of the groups. Hcy increase was related directly to decrease in plasma folate (r = -0.372; P < 0.02). CONCLUSION: These data confirm the beneficial effects of HP diets on body weight but bring attention to the control of folate allowance to limit the adverse effects of elevated Hcy. Ingestion of folate-rich foods or even folate supplementation should be considered when using these HP diets over the long term for weight loss.


Subject(s)
Adipose Tissue/metabolism , Diet, Fat-Restricted , Dietary Proteins/pharmacology , Homocysteine/blood , Hyperhomocysteinemia , Obesity/prevention & control , Weight Gain/drug effects , Animals , Energy Intake , Folic Acid/blood , Hyperhomocysteinemia/metabolism , Male , Rats , Rats, Long-Evans
20.
J Cell Mol Med ; 15(11): 2486-97, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21199330

ABSTRACT

Inflammatory bowel diseases (IBD) result from complex interactions between environmental and genetic factors. Low blood levels of vitamin B12 and folate and genetic variants of related target enzymes are associated with IBD risk, in population studies. To investigate the underlying mechanisms, we evaluated the effects of a methyl-deficient diet (MDD, folate, vitamin B12 and choline) in an experimental model of colitis induced by dextran sodium sulphate (DSS), in rat pups from dams subjected to the MDD during gestation and lactation. Four groups were considered (n = 12-16 per group): C DSS(-) (control/DSS(-)), D DSS(-) (deficient/DSS(-)), C DSS(+) (control/DSS(+)) and D DSS(+) (deficient/DSS(+)). Changes in apoptosis, oxidant stress and pro-inflammatory pathways were studied within colonic mucosa. In rat pups, the MDD produced a decreased plasma concentration of vitamin B12 and folate and an increased homocysteine (7.8 ± 0.9 versus 22.6 ± 1.2 µmol/l, P < 0.001). The DSS-induced colitis was dramatically more severe in the D DSS(+) group compared with each other group, with no change in superoxide dismutase and glutathione peroxidase activity, but decreased expression of caspase-3 and Bax, and increased Bcl-2 levels. The mRNA levels of tumour necrosis factor (TNF)-α and protein levels of p38, cytosolic phospolipase A2 and cyclooxygenase 2 were significantly increased in the D DSS(+) pups and were accompanied by a decrease in the protein level of tissue inhibitor of metalloproteinases (TIMP)3, a negative regulator of TNF-α. MDD may cause an overexpression of pro-inflammatory pathways, indicating an aggravating effect of folate and/or vitamin B12 deficiency in experimental IBD. These findings suggest paying attention to vitamin B12 and folate deficits, frequently reported in IBD patients.


Subject(s)
Choline Deficiency , Colitis, Ulcerative , Folic Acid Deficiency , Vitamin B 12 Deficiency , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/drug effects , Cyclooxygenase 2/biosynthesis , Dextran Sulfate/pharmacology , Diet , Folic Acid/blood , Glutathione Peroxidase/metabolism , Homocysteine/blood , Oxidative Stress/drug effects , Phospholipases A2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Tissue Inhibitor of Metalloproteinase-3/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Vitamin B 12/blood , bcl-2-Associated X Protein/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...