Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Biomolecules ; 14(4)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38672521

ABSTRACT

Rett Syndrome (RTT) is a progressive X-linked neurodevelopmental disorder with no cure. RTT patients show disease-associated symptoms within 18 months of age that include developmental regression, progressive loss of useful hand movements, and breathing difficulties, along with neurological impairments, seizures, tremor, and mental disability. Rett Syndrome is also associated with metabolic abnormalities, and the anti-diabetic drug metformin is suggested to be a potential drug of choice with low or no side-effects. Previously, we showed that in vitro exposure of metformin in a human brain cell line induces MECP2E1 transcripts, the dominant isoform of the MECP2 gene in the brain, mutations in which causes RTT. Here, we report the molecular impact of metformin in mice. Protein analysis of specific brain regions in the male and female mice by immunoblotting indicated that metformin induces MeCP2 in the hippocampus, in a sex-dependent manner. Additional experiments confirm that the regulatory role of metformin on the MeCP2 target "BDNF" is brain region-dependent and sex-specific. Measurement of the ribosomal protein S6 (in both phosphorylated and unphosphorylated forms) confirms the sex-dependent role of metformin in the liver. Our results can help foster a better understanding of the molecular impact of metformin in different brain regions of male and female adult mice, while providing some insight towards its potential in therapeutic strategies for the treatment of Rett Syndrome.


Subject(s)
Hippocampus , Metformin , Methyl-CpG-Binding Protein 2 , Rett Syndrome , Animals , Female , Male , Mice , Brain/metabolism , Brain/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Metformin/pharmacology , Methyl-CpG-Binding Protein 2/drug effects , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects , Rett Syndrome/metabolism , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Ribosomal Protein S6/metabolism , Sex Characteristics , Sex Factors
2.
Addict Biol ; 26(2): e12905, 2021 03.
Article in English | MEDLINE | ID: mdl-32293776

ABSTRACT

Micro-RNAs (miRNAs) are highly evolutionarily conserved short-length/noncoding RNA molecules that modulate a wide range of cellular functions in many cell types by regulating the expression of a variety of targeted genes. miRNAs have also recently emerged as key regulators of neuronal genes mediating the effects of psychostimulant drugs and memory-related neuroplasticity processes. Smoking is a predominant addictive behaviour associated with millions of deaths worldwide, and nicotine is a potent natural psychoactive agonist of cholinergic receptors, highly abundant in cigarettes. The influence of miRNAs modulation on cholinergic signalling in the nervous system remains however poorly explored. Using miRNA knockout mice and biochemical, electrophysiological and pharmacological approaches, we examined the effects of miR-132/212 gene disruption on the levels of hippocampal nicotinic acetylcholine receptors, total ERK and phosphorylated ERK (pERK) and MeCP2 protein levels, and studied the impact of nicotine stimulation on hippocampal synaptic transmission and synaptic depression and strengthening. miR-132/212 deletion significantly altered α7-nAChR and pERK protein levels, but not total ERK or MeCP2, and resulted in both exacerbated synaptic depression and virtually abolished memory-related synaptic strengthening upon nicotine stimulation. These observations reveal a functional miRNAs/nicotinergic signalling interplay critical for nicotinic-receptor expression and neuroplasticity in brain structures relevant for drug addiction and learning and memory functions.


Subject(s)
Dentate Gyrus/drug effects , MicroRNAs/drug effects , Neuronal Plasticity/drug effects , Nicotine/pharmacology , Animals , Extracellular Signal-Regulated MAP Kinases/drug effects , Hippocampus/drug effects , Male , Methyl-CpG-Binding Protein 2/drug effects , Mice , Mice, Knockout , Receptors, Nicotinic/drug effects , Synaptic Transmission/drug effects , eIF-2 Kinase/drug effects
3.
Sci Rep ; 9(1): 2453, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30792426

ABSTRACT

Environmental risk factors contribute to autism spectrum disorders (ASD) etiology. In particular, prenatal exposure to the highly teratogenic anticonvulsant valproic acid (VPA) significantly increases ASD prevalence. Although significant discoveries on the embryopathology of VPA have been reported, its effects on the ability to form enduring social attachment-characteristic of ASD but uncommonly displayed by rats and mice-remains unknown. We aimed to examine the effects of prenatal VPA exposure in the social, monogamous prairie voles (Microtus ochrogaster). Compared to prenatal vehicle-exposed controls, prenatal VPA-exposed prairie voles had lower body weight throughout postnatal development, engaged in fewer social affiliative behaviors in a familial context, exhibited less social interactions with novel conspecifics, and showed enhanced anxiety-like behavior. Along these behavioral deficits, prenatal VPA exposure downregulated prefrontal cortex vasopressin receptor (V1aR) and methyl CpG-binding protein 2 (MeCP2) mRNA expression, but did not alter spine density in adults. Remarkably, adult social bonding behaviors, such as partner preference formation and selective aggression, were not disrupted by prenatal VPA exposure. Collectively, these studies suggest that, in this animal model, VPA alters only certain behavioral domains such as sex-naive anxiety and affiliative behaviors, but does not alter other domains such as social bonding with opposite sex individuals.


Subject(s)
Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Sexual Behavior, Animal/drug effects , Social Behavior , Valproic Acid/pharmacology , Aggression/drug effects , Animals , Anxiety/chemically induced , Anxiety/pathology , Arvicolinae , Behavior, Animal/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Male , Methyl-CpG-Binding Protein 2/drug effects , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Pair Bond , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pregnancy , Receptors, Vasopressin/drug effects , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism
4.
Psychiatry Clin Neurosci ; 72(4): 255-265, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28990703

ABSTRACT

AIM: Early life stress can induce epigenetic changes through genetic and environmental interactions and is a risk factor for depression. Brain-derived neurotrophic factor (BDNF) has been implicated in the pathophysiology of depression and antidepressant drug action. We investigated epigenetic changes at the BDNF exon I promoter in the hippocampus of adult rats subjected to maternal separation (MS) during early life and treated with an antidepressant drug as adults. METHODS: Rat pups were subjected to MS from postnatal day 1 to 21 and received chronic escitalopram (ESC) as adults. We assessed the effects of MS and ESC on BDNF exon I and DNA methyltransferases (DNMT) mRNA levels (quantitative reverse-transcription polymerase chain reaction), acetylated histone H3, and MeCP2 binding to the BDNF promoter I (chromatin immunoprecipitation followed by real-time polymerase chain reaction), and BDNF protein levels (enzyme-linked immunosorbent assay). RESULTS: The levels of BDNF protein, exon I mRNA, histone H3 acetylation, and DNMT1 and DNMT3a mRNA were altered in the MS group compared with the control group. Significant decreases were observed in the BDNF protein, exon I mRNA, and histone H3 acetylation levels and there were significant increases in DNMT1 and DNMT3a mRNA levels. The comparison between the MS + ESC and MS groups revealed significant increases in BDNF protein, exon I mRNA, and histone H3 acetylation levels and significant decreases in MeCP2 and DNMT1 and DNMT3a mRNA levels. CONCLUSION: These findings indicate that MS induced epigenetic changes at the BDNF exon I promoter and these changes were prevented by antidepressant drug treatment during adulthood.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Citalopram/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic/physiology , Hippocampus/metabolism , Histones/metabolism , Maternal Deprivation , Methyl-CpG-Binding Protein 2/metabolism , RNA, Messenger/metabolism , Acetylation , Animals , Brain-Derived Neurotrophic Factor/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/drug effects , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA Methyltransferase 3A , Epigenesis, Genetic/drug effects , Exons , Female , Hippocampus/drug effects , Histones/drug effects , Male , Methyl-CpG-Binding Protein 2/drug effects , Pregnancy , Promoter Regions, Genetic , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley
5.
J Mol Neurosci ; 62(3-4): 309-317, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28616777

ABSTRACT

Oligodendrocytes (OLGs) are the myelinating cells of the central nervous system (CNS), and its proper differentiation is crucial for normal functioning of neurons. Methyl-CpG-binding protein 2 (MeCP2) is a multifunctional methylated DNA binding protein; mutation of which causes Rett syndrome, a severe neurodevelopmental disorder. Previously, we reported that MeCP2 is expressed in all the stages of oligodendrocyte development, and also shown the role of MeCP2 as a transcription regulator of myelin genes in OLGs. The expression and function of MeCP2 phosphorylation at S80 (pS80MeCP2) has been well studied in neurons and astrocytes; however, there is no data so far available in OLGs regarding pS80MeCP2. Certain developmental stimuli such as growth factors and extracellular matrix (ECM) protein play important role in OLG development. In the present study, we have examined the effects of external stimuli (growth factors (GFs) and extracellular matrix (ECMs)) on S80 phosphorylation of MeCP2 in N19 oligodendroglial cells (N19 OLGs). This study provides the first evidence that laminin (LN) differentially regulates the expression of pS80MeCP2 in immature and mature N19 OLGs. Thus, MeCP2 is phosphorylated in a stimulus-dependent manner during oligodendrocyte development, and thereby, it may regulate the oligodendrocyte behavior.


Subject(s)
Laminin/pharmacology , Methyl-CpG-Binding Protein 2/metabolism , Oligodendroglia/metabolism , Protein Processing, Post-Translational , Animals , Cell Line , Methyl-CpG-Binding Protein 2/drug effects , Methyl-CpG-Binding Protein 2/genetics , Mice , Phosphorylation
6.
Psychopharmacology (Berl) ; 234(6): 913-923, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28116477

ABSTRACT

RATIONALE: Phosphorylation of the methyl DNA-binding protein MeCP2 at Ser421 (pMeCP2-S421) is induced in corticolimbic brain regions during exposure to drugs of abuse and modulates reward-driven behaviors. However, whether pMeCP2-S421 is also involved in behavioral adaptations to aversive drugs is unknown. OBJECTIVES: Our goal was to establish the role and regulation of pMeCP2-S421 in corticolimbic brain regions of mice upon acute treatment with the kappa opioid receptor agonist U50488 and during the expression of U50488-induced conditioned place aversion (CPA). METHODS: pMeCP2-S421 levels were measured in the nucleus accumbens (NAc), prelimbic cortex, infralimbic cortex (ILC), and basolateral amygdala (BLA) of male mice after intraperitoneal administration of U50488 and upon the expression of U50488-induced CPA. Fos was measured as marker of neural activity in the same brain regions. U50488-induced CPA and Fos levels were compared between knockin (KI) mice that lack pMeCP2-S421 and their wild-type (WT) littermates. RESULTS: U50488 administration acutely induced pMeCP2-S421 and Fos selectively in the NAc but did not alter MeCP2 levels in any brain region. U50488-induced CPA was associated with decreased pMeCP2-S421 in the ILC and BLA and induced Fos in the BLA. MeCP2 KI mice showed CPA indistinguishable from their WT littermates, but they also showed less BLA Fos induction upon CPA. CONCLUSION: These data are the first to show that pMeCP2-S421 is induced in the brain acutely after U50488 administration but not upon U50488-induced CPA. Although pMeCP2-S421 is not required for U50488-induced CPA, this phosphorylation event may contribute to molecular plasticities in brain regions that govern aversive behaviors.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Brain/drug effects , Conditioning, Psychological/drug effects , Methyl-CpG-Binding Protein 2/drug effects , Receptors, Opioid, kappa/agonists , Animals , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Behavior, Animal/drug effects , Brain/metabolism , Gene Knock-In Techniques , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Reward
7.
Addict Biol ; 22(2): 369-380, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26610727

ABSTRACT

Alcohol use disorder is the outcome of both genetic and environmental influences and their interaction via epigenetic mechanisms. The neurotransmitter glutamate is an important regulator of reward circuits and implicated in adaptive changes induced by ethanol intake. The present study aimed at investigating corticolimbic and corticostriatal genetic signatures focusing on the glutamatergic phenotype in relation to early-life stress (ELS) and consequent adult ethanol consumption. A rodent maternal separation model was employed to mimic ELS, and a free-choice paradigm was used to assess ethanol intake in adulthood. Gene expression levels of the Vesicular Glutamate Transporters (Vglut) 1, 2 and 3, as well as two key regulators of DNA methylation, DNA (cytosine-5)-methyltransferase 1 (Dnmt1) and methyl-CpG-binding protein 2 (Mecp2), were analyzed. Brain regions of interest were the ventral tegmental area (VTA), nucleus accumbens (Acb), medial prefrontal cortex (mPFC) and dorsal striatum (dStr), all involved in mediating aspects of ethanol reward. Region-specific Vglut, Dnmt1 and Mecp2 expression patterns were observed. ELS was associated with down-regulated expression of Vglut2 in the VTA and mPFC. Rats exposed to ELS were more sensitive to ethanol-induced changes in Vglut expression in the VTA, Acb, and dStr and in Dnmt1 and Mecp2 expression in the striatal regions. These findings suggest long-term glutamatergic and DNA methylation neuroadaptations as a consequence of ELS, and show an association between voluntary drinking in non-preferring, non-dependent, rodents and different Vglut, Dnmt1 and Mecp2 expression depending on early-life history.


Subject(s)
Central Nervous System Depressants/pharmacology , Corpus Striatum/drug effects , DNA Methylation/drug effects , Ethanol/pharmacology , Gene Expression/drug effects , Limbic System/drug effects , Maternal Deprivation , Presynaptic Terminals/drug effects , Stress, Psychological/genetics , Animals , Brain/drug effects , Brain/metabolism , Choice Behavior , Corpus Striatum/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Female , Glutamic Acid/metabolism , Limbic System/metabolism , Male , Methyl-CpG-Binding Protein 2/drug effects , Methyl-CpG-Binding Protein 2/genetics , Neostriatum/drug effects , Neostriatum/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Presynaptic Terminals/metabolism , Rats , Rats, Wistar , Reward , Stress, Psychological/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Vesicular Glutamate Transport Protein 1/drug effects , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 2/drug effects , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Proteins/drug effects , Vesicular Glutamate Transport Proteins/genetics
8.
Psychopharmacology (Berl) ; 233(17): 3279-88, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27392631

ABSTRACT

RATIONALE: Early life stress is a major risk factor for cocaine addiction; however, the underlying molecular mechanisms remain relatively unexplored. MicroRNA-212 (miR-212) and methyl CpG binding protein 2 (MeCP2) have recently emerged as key regulators of brain-derived neurotrophic factor (BDNF) signaling during the acquisition and maintenance of cocaine-seeking behaviors. OBJECTIVES: We therefore investigated the effect of maternal separation (MS) on cocaine-induced conditioned place preference (CPP) during periadolescence and how this influences miR-212, Mecp2, and Bdnf expressions in the prefrontal cortex. METHODS: Male BALB/c mice subjected to MS (3 h/day) from postnatal day 2 to 15 or normal animal facility rearing (AFR) were tested for CPP at postnatal day 45, or not exposed to experimental manipulations (drug-naïve animals). Cultured primary cortical neurons were used to determine miR-212 expression changes following depolarization by KCL treatment. RESULTS: MS increased cocaine-induced CPP and decreased Bdnf exon IV expression, which correlated with higher CPP scores in such animals. An experience-dependent decrease in miR-212 expression was observed following CPP test. This effect was mimicked in primary cortical neurons in vitro, under activity-dependent conditions. In contrast, increased Mecp2 expression was found after CPP test, suggesting an opposing relationship between miR-212 and Mecp2 expression following cocaine place preference acquisition. However, these effects were not present in mice exposed to MS. CONCLUSIONS: Together, our results suggest that early life stress can enhance the motivational salience for cocaine-paired cues during periadolescence, and that altered expression of miR-212, Mecp2, and Bdnf in the prefrontal cortex is involved in this process.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cocaine/pharmacology , Conditioning, Classical/drug effects , Dopamine Uptake Inhibitors/pharmacology , Maternal Deprivation , Methyl-CpG-Binding Protein 2/genetics , MicroRNAs/metabolism , Prefrontal Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/drug effects , Cocaine-Related Disorders , Male , Methyl-CpG-Binding Protein 2/drug effects , Mice , Mice, Inbred BALB C , MicroRNAs/drug effects , Prefrontal Cortex/drug effects
9.
Invest Ophthalmol Vis Sci ; 56(9): 5579-89, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26305530

ABSTRACT

PURPOSE: The purpose of this study was to evaluate expression of methyl-CpG-binding protein 2 (MeCP2) in epiretinal membranes from patients with proliferative vitreoretinopathy (PVR) and to investigate effects of inhibition of MeCP2 and DNA methylation on transforming growth factor (TGF)-ß-induced retinal pigment epithelial (RPE) cell transdifferentiation. METHODS: Expression of MeCP2 and its colocalization with cytokeratin and α-smooth muscle actin (α-SMA) in surgically excised PVR membranes was studied using immunohistochemistry. The effects of 5-AZA-2'-deoxycytidine (5-AZA-dC) on human RPE cell migration and viability were evaluated using a modified Boyden chamber assay and the colorimetric 3-(4,5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide (MTT) assay. Expression of RASAL1 mRNA and its promoter region methylation were evaluated by real-time PCR and methylation-specific PCR. Effects of 5-AZA-dC on expression of α-SMA, fibronectin (FN), and TGF-ß receptor 2 (TGF-ß R2) and Smad2/3 phosphorylation were analyzed by Western blotting. Effect of short interfering RNA (siRNA) knock-down of MeCP2 on expression of α-SMA and FN induced by TGFß was determined. RESULTS: MeCP2 was abundantly expressed in cells within PVR membranes where it was double labeled with cells positive for cytokeratin and α-SMA. 5-AZA-dC inhibited expression of MeCP2 and suppressed RASAL1 gene methylation while increasing expression of the RASAL1 gene. Treatment with 5-AZA-dC significantly suppressed the expression of α-SMA, FN, TGF-ß R2 and phosphorylation of Smad2/3 and inhibited RPE cell migration. TGF-ß induced expression of α-SMA, and FN was suppressed by knock-down of MeCP2. CONCLUSIONS: MeCP2 and DNA methylation regulate RPE transdifferentiation and may be involved in the pathogenesis of PVR.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , DNA/genetics , Gene Expression Regulation , Methyl-CpG-Binding Protein 2/genetics , Retinal Pigment Epithelium/metabolism , Vitreoretinopathy, Proliferative/genetics , Azacitidine/pharmacology , Blotting, Western , Cell Movement , Cell Transdifferentiation , Cells, Cultured , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunohistochemistry , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/drug effects , Phosphorylation , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Vitreoretinopathy, Proliferative/drug therapy , Vitreoretinopathy, Proliferative/metabolism
10.
Curr Alzheimer Res ; 9(5): 555-62, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22272629

ABSTRACT

Late onset Alzheimer's disease (LOAD) is typical of the majority of Alzheimer's disease (AD) cases (~90%), and has no clear genetic association. Previous studies from our lab suggest that an epigenetic component could be involved. Developmental exposure of primates and rodents to lead (Pb) predetermined the expression of AD-related genes, such as the amyloid-ß precursor protein (AßPP), later in life. In addition to AßPP, the preponderance of genes that were reprogrammed was rich in CpG dinucleotides implicating DNA methylation and chromatin restructuring in their regulation. To examine the involvement of epigenetic intermediates in Pb-induced alterations in gene expression, differentiated SH-SY5Y cells were exposed to a series of Pb concentrations (5-100 µM) for 48 h and were analyzed for the protein expression of AßPP, ß-site amyloid precursor protein cleaving enzyme 1 (BACE1), specificity protein 1 and 3 (Sp1, Sp3) and epigenetic intermediates like DNA methyltransferase 1, 3a (Dnmt1, Dnmt3a) and methyl CpG binding protein 2 (MeCP2) involved in DNA methylation six days after the exposure had ceased. Western blot analysis indicated a significant latent elevation in AD biomarkers as well as the transcription factors Sp1 and Sp3, accompanied by a significant reduction in the protein levels of DNA methylating enzymes. RT-PCR analysis of Dnmt1, Dnmt3a and MeCP2 indicated a significant down-regulation of the mRNA levels. These data suggest that Pb interferes with DNA methylating capacity in these cells, thus altering the expression of AD-related genes.


Subject(s)
Alzheimer Disease/etiology , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Lead/toxicity , Nerve Tissue Proteins/drug effects , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Biomarkers/metabolism , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Environmental Exposure , Epigenesis, Genetic/drug effects , Humans , Methyl-CpG-Binding Protein 2/drug effects , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/analysis , Sp Transcription Factors/drug effects , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL