Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Neuropharmacology ; 131: 271-281, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29223527

ABSTRACT

Adverse maternal behaviors during pregnancy and unfavorable postnatal experiences during development are associated with an increased risk of developing psychiatric disorders, as well as, a vulnerability to alcohol addiction in adulthood. Here, we examined the effects of combined ethanol exposure during late pregnancy and postnatal maternal separation (MS) on HPA responsiveness, anxiety behavior and preference for alcohol consumption in adult male rats. Animals exposed to both conditions revealed a decrease in blood levels of allopregnanolone accompanied by increased anxiety behavior. In addition, basal blood levels of corticosterone were markedly decreased in all experimental groups while increases in the foot-shock-induced corticosterone levels were more pronounced in MS animals. Finally, evaluating EtOH drinking behavior, MS animals exhibited a remarkable EtOH preference even at low doses (0.1-1%). Altogether, these data suggest that adverse conditions, alone or in combination, may alter anxiety-like states as well as modify behavior towards alcohol consumption.


Subject(s)
Alcohol Drinking/metabolism , Anxiety/metabolism , Corticosterone/blood , Fetal Alcohol Spectrum Disorders/metabolism , Maternal Deprivation , Pregnanolone/blood , Alcohol Drinking/psychology , Analysis of Variance , Animals , Anxiety/etiology , Electroshock , Fetal Alcohol Spectrum Disorders/psychology , Male , Random Allocation , Rats, Sprague-Dawley , Stress, Psychological/metabolism
2.
Transl Psychiatry ; 6: e711, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26756904

ABSTRACT

We have recently reported that mice born from dams stressed during pregnancy (PRS mice), in adulthood, have behavioral deficits reminiscent of behaviors observed in schizophrenia (SZ) and bipolar (BP) disorder patients. Furthermore, we have shown that the frontal cortex (FC) and hippocampus of adult PRS mice, like that of postmortem chronic SZ patients, are characterized by increases in DNA-methyltransferase 1 (DNMT1), ten-eleven methylcytosine dioxygenase 1 (TET1) and exhibit an enrichment of 5-methylcytosine (5MC) and 5-hydroxymethylcytosine (5HMC) at neocortical GABAergic and glutamatergic gene promoters. Here, we show that the behavioral deficits and the increased 5MC and 5HMC at glutamic acid decarboxylase 67 (Gad1), reelin (Reln) and brain-derived neurotrophic factor (Bdnf) promoters and the reduced expression of the messenger RNAs (mRNAs) and proteins corresponding to these genes in FC of adult PRS mice is reversed by treatment with clozapine (5 mg kg(-1) twice a day for 5 days) but not by haloperidol (1 mg kg(-1) twice a day for 5 days). Interestingly, clozapine had no effect on either the behavior, promoter methylation or the expression of these mRNAs and proteins when administered to offspring of nonstressed pregnant mice. Clozapine, but not haloperidol, reduced the elevated levels of DNMT1 and TET1, as well as the elevated levels of DNMT1 binding to Gad1, Reln and Bdnf promoters in PRS mice suggesting that clozapine, unlike haloperidol, may limit DNA methylation by interfering with DNA methylation dynamics. We conclude that the PRS mouse model may be useful preclinically in screening for the potential efficacy of antipsychotic drugs acting on altered epigenetic mechanisms. Furthermore, PRS mice may be invaluable for understanding the etiopathogenesis of SZ and BP disorder and for predicting treatment responses at early stages of the illness allowing for early detection and remedial intervention.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Chromatin Assembly and Disassembly/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Stress, Psychological/physiopathology , Animals , Behavior, Animal/physiology , Blotting, Western , Brain/physiopathology , Chromatin Assembly and Disassembly/physiology , Clozapine/pharmacology , Disease Models, Animal , Epigenesis, Genetic/drug effects , Female , Mice , Pregnancy , Real-Time Polymerase Chain Reaction , Reelin Protein
3.
Schizophr Res ; 167(1-3): 35-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25476119

ABSTRACT

The down regulation of glutamic acid decarboxylase67 (GAD1), reelin (RELN), and BDNF expression in brain of schizophrenia (SZ) and bipolar (BP) disorder patients is associated with overexpression of DNA methyltransferase1 (DNMT1) and ten-eleven translocase methylcytosine dioxygenase1 (TET1). DNMT1 and TET1 belong to families of enzymes that methylate and hydroxymethylate cytosines located proximal to and within cytosine phosphodiester guanine (CpG) islands of many gene promoters, respectively. Altered promoter methylation may be one mechanism underlying the down-regulation of GABAergic and glutamatergic gene expression. However, recent reports suggest that both DNMT1 and TET1 directly bind to unmethylated CpG rich promoters through their respective Zinc Finger (ZF-CXXC) domains. We report here, that the binding of DNMT1 to GABAergic (GAD1, RELN) and glutamatergic (BDNF-IX) promoters is increased in SZ and BP disorder patients and this increase does not necessarily correlate with enrichment in promoter methylation. The increased DNMT1 binding to these promoter regions is detected in the cortex but not in the cerebellum of SZ and BP disorder patients, suggesting a brain region and neuron specific dependent mechanism. Increased binding of DNMT1 positively correlates with increased expression of DNMT1 and with increased binding of MBD2. In contrast, the binding of TET1 to RELN, GAD1 and BDNF-IX promoters failed to change. These data are consistent with the hypothesis that the down-regulation of specific GABAergic and glutamatergic genes in SZ and BP disorder patients may be mediated, at least in part, by a brain region specific and neuronal-activity dependent DNMT1 action that is likely independent of its DNA methylation activity.


Subject(s)
Bipolar Disorder/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Schizophrenia/pathology , gamma-Aminobutyric Acid/metabolism , Aged , Aged, 80 and over , Analysis of Variance , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Chromatin Immunoprecipitation , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , Male , Middle Aged , Mixed Function Oxygenases , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/physiology , Protein Binding/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
4.
J Neurogenet ; 28(1-2): 41-52, 2014.
Article in English | MEDLINE | ID: mdl-24702539

ABSTRACT

Schizophrenia (SZ) is a heritable, nonmendelian, neurodevelopmental disorder in which epigenetic dysregulation of the brain genome plays a fundamental role in mediating the clinical manifestations and course of the disease. The authors recently reported that two enzymes that belong to the dynamic DNA methylation/demethylation network-DNMT (DNA methyltransferase) and TET (ten-eleven translocase; 5-hydroxycytosine translocator)-are abnormally increased in corticolimbic structures of SZ postmortem brain, suggesting a causal relationship between clinical manifestations of SZ and changes in DNA methylation and in the expression of SZ candidate genes (e.g., brain-derived neurotrophic factor [BDNF], glucocorticoid receptor [GCR], glutamic acid decarboxylase 67 [GAD67], reelin). Because the clinical manifestations of SZ typically begin with a prodrome followed by a first episode in adolescence with subsequent deterioration, it is obvious that the natural history of this disease cannot be studied only in postmortem brain. Hence, the focus is currently shifting towards the feasibility of studying epigenetic molecular signatures of SZ in blood cells. Initial studies show a significant enrichment of epigenetic changes in lymphocytes in gene networks directly relevant to psychiatric disorders. Furthermore, the expression of DNA-methylating/demethylating enzymes and SZ candidate genes such as BDNF and GCR are altered in the same direction in both brain and blood lymphocytes. The coincidence of these changes in lymphocytes and brain supports the hypothesis that common environmental or genetic risk factors are operative in altering the epigenetic components involved in orchestrating transcription of specific genes in brain and peripheral tissues. The identification of DNA methylation signatures for SZ in peripheral blood cells of subjects with genetic and clinical high risk would clearly have potential for the diagnosis of SZ early in its course and would be invaluable for initiating early intervention and individualized treatment plans.


Subject(s)
Biomarkers/blood , Epigenesis, Genetic/genetics , Lymphocytes/metabolism , Schizophrenia , DNA Methylation , Gene Regulatory Networks , Humans , Reelin Protein , Schizophrenia/blood , Schizophrenia/genetics , Schizophrenia/metabolism
5.
Transl Psychiatry ; 4: e349, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24448211

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by symptoms related to altered social interactions/communication and restricted and repetitive behaviors. In addition to genetic risk, epigenetic mechanisms (which include DNA methylation/demethylation) are thought to be important in the etiopathogenesis of ASD. We studied epigenetic mechanisms underlying the transcriptional regulation of candidate genes in cerebella of ASD patients, including the binding of MeCP2 (methyl CpG binding protein-2) to the glutamic acid decarboxylase 67 (GAD1), glutamic acid decarboxylase 65 (GAD2), and Reelin (RELN) promoters and gene bodies. Moreover, we performed methyl DNA immunoprecipitation (MeDIP) and hydroxymethyl DNA immunoprecipitation (hMeDIP) to measure total 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) in the same regions of these genes. The enrichment of 5-hmC and decrease in 5-mC at the GAD1 or RELN promoters detected by 5-hmC and 5-mC antibodies was confirmed by Tet-assisted bisulfite (TAB) pyrosequencing. The results showed a marked and significant increase in MeCP2 binding to the promoter regions of GAD1 and RELN, but not to the corresponding gene body regions in cerebellar cortex of ASD patients. Moreover, we detected a significant increase in TET1 expression and an enrichment in the level of 5-hmC, but not 5-mC, at the promoters of GAD1 and RELN in ASD when compared with CON. Moreover, there was increased TET1 binding to these promoter regions. These data are consistent with the hypothesis that an increase of 5-hmC (relative to 5-mC) at specific gene domains enhances the binding of MeCP2 to 5-hmC and reduces expression of the corresponding target genes in ASD cerebella.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebellar Cortex/metabolism , Child Development Disorders, Pervasive/metabolism , Cytosine/analogs & derivatives , Extracellular Matrix Proteins/metabolism , Glutamate Decarboxylase/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Tissue Banks , 5-Methylcytosine/analogs & derivatives , Cell Adhesion Molecules, Neuronal/genetics , Cerebellar Cortex/pathology , Child Development Disorders, Pervasive/genetics , Cytosine/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Glutamate Decarboxylase/genetics , Humans , Mixed Function Oxygenases , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Reelin Protein , Serine Endopeptidases/genetics
6.
Neuropharmacology ; 60(7-8): 1007-16, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21074545

ABSTRACT

It is becoming increasingly clear that a dysfunction of the GABAergic/glutamatergic network in telencephalic brain structures may be the pathogenetic mechanism underlying psychotic symptoms in schizophrenia (SZ) and bipolar (BP) disorder patients. Data obtained in Costa's laboratory (1996-2009) suggest that this dysfunction may be mediated primarily by a downregulation in the expression of GABAergic genes (e.g., glutamic acid decarboxylase67[GAD67] and reelin) associated with DNA methyltransferase (DNMT)-dependent hypermethylation of their promoters. A pharmacological strategy to reduce the hypermethylation of GABAergic promoters is to administer drugs, such as the histone deacetylase (HDAC) inhibitor valproate (VPA), that induce DNA-demethylation when administered at doses that facilitate chromatin remodeling. The benefits elicited by combining VPA with antipsychotics in the treatment of BP disorder suggest that an investigation of the epigenetic interaction of these drugs is warranted. Our studies in mice suggest that when associated with VPA, clinically relevant doses of clozapine elicit a synergistic potentiation of VPA-induced GABAergic promoter demethylation. Olanzapine and quetiapine (two clozapine congeners) also facilitate chromatin remodeling but at doses higher than used clinically, whereas haloperidol and risperidone are inactive. Hence, the synergistic potentiation of VPA's action on chromatin remodeling by clozapine appears to be a unique property of the dibenzepines and is independent of their action on catecholamine or serotonin receptors. By activating DNA-demethylation, the association of clozapine or its derivatives with VPA or other more potent and selective HDAC inhibitors may be considered a promising treatment strategy for normalizing GABAergic promoter hypermethylation and the GABAergic gene expression downregulation detected in the postmortem brain of SZ and BP disorder patients. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.


Subject(s)
Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Epigenesis, Genetic/drug effects , Schizophrenia/drug therapy , gamma-Aminobutyric Acid/genetics , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacology , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Excitatory Amino Acid Agents/metabolism , Excitatory Amino Acid Agents/pharmacology , Excitatory Amino Acid Agents/therapeutic use , Gene Expression/drug effects , Humans , Interneurons/drug effects , Interneurons/physiology , Mice , Molecular Targeted Therapy , Reelin Protein , Schizophrenia/genetics , Schizophrenia/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Proc Natl Acad Sci U S A ; 105(36): 13614-9, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18757738

ABSTRACT

Cortical GABAergic dysfunction, a hallmark of both schizophrenia (SZ) and bipolar (BP) disorder pathophysiologies may relate to the hypermethylation of GABAergic gene promoters (i.e., reelin and GAD67). Benefits elicited by a combination of atypical antipsychotics with valproate (VPA) (a histone deacetylase inhibitor that may also activate brain DNA demethylation) in SZ or BP disorder treatment prompted us to investigate whether the beneficial action of this association depends on induction of a putative DNA demethylase activity. To monitor this activity, we measured the ratio of 5-methyl cytosine to unmethylated cytosine in reelin and GAD67 promoters in the mouse frontal cortex and striatum. We compared normal mice with mice pretreated with l-methionine (5.2 mmol/kg s.c. twice a day for 7 days) to hypermethylate promoters, including reelin and GAD67. Clinically relevant doses of clozapine (CLZ) (3.8 to 15 micromol/kg twice a day s.c. for 3 days) and sulpiride (SULP) (12.5 to 50 micromol/kg twice a day for 3 days) but not clinically relevant doses of haloperidol (HAL) (1.3 to 4 micromol/kg twice a day s.c. for 3 days) or olanzapine (OLZ) (4 to 15 micromol/kg twice a day for 3 days) exhibited dose-related increases in the cortical and striatal demethylation of hypermethylated reelin and GAD67 promoters. These effects of CLZ and SULP were dramatically potentiated by a clinically relevant VPA dose (0.5 mmol/kg twice a day for 3 days). By activating a DNA demethylase, the association of CLZ or SULP with VPA may facilitate a chromatin remodeling that normalizes the GABAergic gene expression down-regulation detected in the telencephalic regions of SZ and BP patients.


Subject(s)
Brain/drug effects , Brain/metabolism , Clozapine/pharmacology , DNA Methylation , Sulpiride/pharmacology , Acetylation , Animals , Benzodiazepines/pharmacology , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , DNA Methylation/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Haloperidol/pharmacology , Histones/metabolism , Lysine/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Olanzapine , Promoter Regions, Genetic/genetics , Reelin Protein , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Valproic Acid/pharmacology
8.
Proc Natl Acad Sci U S A ; 104(11): 4676-81, 2007 Mar 13.
Article in English | MEDLINE | ID: mdl-17360583

ABSTRACT

Reelin and glutamic acid decarboxylase 67 (GAD(67)) expression down-regulation in GABAergic interneurons of mice exposed to protracted treatment with l-methionine (MET) is attributed to RELN and GAD(67) promoter cytosine-5-hypermethylation. This process recruits various transcription repressor proteins [methyl-CpG binding protein (MeCP2) and histone deacetylases (HDACs)] leading to formation of transcriptionally inactive chromatin. Here, we tested the hypothesis that RELN and GAD(67) promoter cytosine-5-hypermethylation induced by a protracted MET treatment is reversible and that repeated administration of HDAC inhibitors influences this process by an activation of DNA-cytosine-5-demethylation. In the frontal cortices of mice receiving MET (5.2 mmol/kg twice a day for 7 days) and killed at 1, 2, 3, 6, and 9 days during MET washout, we measured RELN (base pairs -414 to -242) and GAD(67) (base pairs -1133 to -942) promoter methylation and MeCP2 bound to methylated cytosines of RELN (base pairs -520 to -198) and GAD(67) (base pairs -446 to -760) promoters. Levels of RELN and GAD(67) promoter hypermethylation induced by 7 days of MET treatment declines by approximately 50% after 6 days of MET withdrawal. When valproate (VPA) (2 mmol/kg) or MS-275 (0.015-0.12 mmol/kg), two structurally unrelated HDAC inhibitors, was given after MET treatment termination, VPA and MS-275 dramatically accelerated RELN and GAD(67) promoter demethylation in 48-72 h. At these doses, VPA and MS-275 effectively increased the binding of acetylhistone-3 to RELN and GAD(67) promoters, suggesting that histone-3 covalent modifications modulate DNA demethylation in terminally differentiated neurons, supporting the view that, directly or indirectly, HDAC inhibitors may facilitate DNA demethylation.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Glutamate Decarboxylase/chemistry , Glutamate Decarboxylase/genetics , Histones/chemistry , Isoenzymes/chemistry , Isoenzymes/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Acetylation , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cytosine/chemistry , DNA Methylation , Down-Regulation , Extracellular Matrix Proteins/metabolism , Male , Methylation , Mice , Nerve Tissue Proteins/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Time Factors , Valproic Acid/pharmacology
9.
Mol Psychiatry ; 12(4): 385-97, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17264840

ABSTRACT

Among the most consistent results of studies of post-mortem brain tissue from schizophrenia patients (SZP) is the finding that in this disease, several genes expressed by GABAergic neurons are downregulated. This downregulation may be caused by hypermethylation of the relevant promoters in affected neurons. Indeed, increased numbers of GABAergic interneurons expressing DNA methyltransferase 1 (DNMT1) mRNA have been demonstrated in the prefrontal cortex (PFC) of SZP using in situ hybridization. The present study expands upon these findings using nested competitive reverse transcription-polymerase chain reaction combined with laser-assisted microdissection to quantitate the extent of DNMT1 mRNA overexpression in distinct populations of GABAergic neurons obtained from either layer I or layer V of the PFC of SZP. In a cohort of eight SZP and eight non-psychiatric subject (NPS) post-mortem BA9 tissue samples, DNMT1 mRNA was found to be selectively expressed in GABAergic interneurons and virtually absent in pyramidal neurons. DNMT1 mRNA expression was approximately threefold higher in GABAergic interneurons microdissected from layer I of SZP relative to the same neurons microdissected from NPS. GABAergic interneurons obtained from layer V of the same samples displayed no difference in DNMT1 mRNA expression between groups. In the same samples, the GABAergic neuron-specific glutamic acid-decarboxylase(67) (GAD(67)) and reelin mRNAs were underexpressed twofold in GABAergic interneurons isolated from layer I of SZP relative to GABAergic interneurons microdissected from layer I of NPS, and unaltered in GABAergic interneurons of layer V. These findings implicate an epigenetically mediated layer I GABAergic dysfunction in the pathogenesis of schizophrenia, and suggest novel strategies for treatment of the disease.


Subject(s)
Epigenesis, Genetic/physiology , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/pathology , Schizophrenia , gamma-Aminobutyric Acid/metabolism , Adult , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Glutamate Decarboxylase/metabolism , Humans , Isoenzymes/metabolism , Male , Microdissection/methods , Middle Aged , RNA, Messenger/biosynthesis , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction/methods , Schizophrenia/etiology , Schizophrenia/genetics , Schizophrenia/pathology
11.
Proc Natl Acad Sci U S A ; 102(35): 12578-83, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16113080

ABSTRACT

Reduction of prefrontal cortex glutamic acid decarboxylase (GAD67) and reelin (mRNAs and proteins) expression is the most consistent finding reported by several studies of postmortem schizophrenia (SZ) brains. Converging evidence suggests that the reduced GAD67 and reelin expression in cortical GABAergic interneurons of SZ brains is the consequence of an epigenetic hypermethylation of RELN and GAD67 promoters very likely mediated by the overexpression of DNA methyltransferase 1 in cortical GABAergic interneurons. Studies of the molecular mechanisms (DNA methylation plus related chromatin remodeling factors) that cause the down-regulation of reelin and GAD67 in SZ brains have important implications not only to understand the disease pathogenesis but also to improve present pharmacological interventions to treat SZ. The mouse treated with l-methionine models some of the molecular neuropathologies detected in SZ, including the hypermethylation of RELN promoter CpG islands and the down-regulation of reelin and GAD67 expression. We now report that in these mice, RELN and GAD67 promoters express an increased recruitment of methyl-CpG binding domain proteins. In these mice the histone deacetylase inhibitor valproate, which increases acetylated histone content in cortical GABAergic interneurons, also prevents MET-induced RELN promoter hypermethylation and reduces the methyl-CpG binding domain protein binding to RELN and GAD67 promoters. These findings suggest that DNA hypermethylation and the associated chromatin remodeling may be critically important in mediating the epigenetic down-regulation of reelin and GAD67 expression detected in cortical GABAergic interneurons of SZ patients.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Glutamate Decarboxylase/genetics , Isoenzymes/genetics , Nerve Tissue Proteins/genetics , Schizophrenia/genetics , Serine Endopeptidases/genetics , Animals , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA/genetics , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Down-Regulation , Epigenesis, Genetic , Gene Expression , Globins/genetics , Methionine/toxicity , Methyl-CpG-Binding Protein 2 , Mice , Models, Biological , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reelin Protein , Repressor Proteins/metabolism , Schizophrenia/chemically induced , Schizophrenia/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Crit Rev Neurobiol ; 16(1-2): 1-23, 2004.
Article in English | MEDLINE | ID: mdl-15581395

ABSTRACT

Several lines of evidence support the role of an epigenetic-induced GABAergic cortical dysfunction in schizophrenia psychopathology, which is probably dependent on an increase in the expression of DNA-methyltransferase-1 occurring selectively in GABAergic neurons. The key enzyme regulating GABA synthesis, termed glutamic acid decarboxylase 67 (GAD67) and the important neurodevelopmental protein called reelin are coexpressed in GABAergic neurons. Upon release, GABA and reelin bind to postsynaptic receptors located in dendrites, somata, or the axon initial segment of pyramidal neurons. Because GAD67 and reelin are downregulated in schizophrenia, it is suggested that schizophrenics may express GABAergic deficit-related alterations of pyramidal neuron function. A reduction of dendritic spines is a finding reported in the prefrontal cortex of schizophrenia patients. Because dendritic spines are innervated by glutamatergic axon terminals, very probably this reduction of dendritic spine expression is translated into a functional deficit of glutamatergic transmission. Plastic modifications of neuronal circuits are probably dependent on GABAergic transmitter tone, and it is likely that GABAergic dysfunction is at the root of synaptic plasticity deficits in schizophrenia. Thus, a possible avenue for the treatment of schizophrenia would be to address this GABAergic functional deficit using positive allosteric modulators of the action of GABA at GABAA receptors. Benzodiazepines (BZ) such as diazepam are effective in treating positive and negative symptoms of schizophrenia, but because they positively modulate GABAA receptors expressing alpha1 subunits, these BZs cause sedation and tolerance. In contrast, imidazenil, a full allosteric modulator of GABAA receptors expressing alpha5 subunits may reduce psychotic symptomatology without producing sedation. Hence, imidazenil should be appropriately studied as a prospective candidate for a pharmacological intervention in schizophrenia.


Subject(s)
Cerebral Cortex/physiopathology , Schizophrenia/physiopathology , gamma-Aminobutyric Acid/physiology , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Cerebral Cortex/pathology , Dendrites/pathology , Dendrites/physiology , Excitatory Amino Acid Antagonists/pharmacology , Hallucinogens/pharmacology , Humans , Neuronal Plasticity/physiology , Neurons/pathology , Phencyclidine/pharmacology , Pyramidal Cells/pathology , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Reelin Protein , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/pathology
14.
Proc Natl Acad Sci U S A ; 101(1): 348-53, 2004 Jan 06.
Article in English | MEDLINE | ID: mdl-14684836

ABSTRACT

A down-regulation of reelin and glutamic acid decarboxylase (GAD) 67 mRNAs was detected in gamma-aminobutyric acid (GABA)ergic cortical interneurons of schizophrenia (SZ) postmortem brains (10), suggesting that the availability of GABA and reelin may be decreased in SZ cortex. In situ hybridization of the mRNA encoding for DNA-methyltransferase 1, which catalyzes the methylation of promoter CpG islands, shows that the expression of this mRNA is increased in cortical GABAergic interneurons but not in pyramidal neurons of SZ brains. Counts of reelin mRNA-positive neurons in Brodmann's area 10 of either nonpsychiatric subjects or SZ patients show that the expression of reelin mRNA is decreased in layer-I, -II, and -IV GABAergic interneurons of SZ patients. These findings are consistent with the hypothesis that the increase of DNA-methyltransferase 1 expression in telencephalic GABAergic interneurons of SZ patients causes a promoter hypermethylation of reelin and GAD(67) and perhaps of other genes expressed in these interneurons. It is difficult to decide whether this dysfunction of GABAergic neurons detected in SZ is responsible for this disease or is a consequence of this disorder. Although at present we cannot differentiate between these two alternatives, it is important to consider that so far a molecular pathology of cortical GABAergic neurons appears to be the most consistent finding associated with SZ morbidity.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizophrenia/enzymology , Schizophrenia/genetics , Telencephalon/enzymology , Adult , Aged , Case-Control Studies , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Gene Expression , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , Interneurons/enzymology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Middle Aged , Nerve Tissue Proteins , Promoter Regions, Genetic , Reelin Protein , Serine Endopeptidases , gamma-Aminobutyric Acid/metabolism
15.
Proc Natl Acad Sci U S A ; 99(26): 17095-100, 2002 Dec 24.
Article in English | MEDLINE | ID: mdl-12481028

ABSTRACT

Reelin and glutamic acid decarboxylase (GAD)67 expressed by cortical gamma-aminobutyric acid-ergic interneurons are down-regulated in schizophrenia. Because epidemiological studies of schizophrenia fail to support candidate gene haploinsufficiency of Mendelian origin, we hypothesize that epigenetic mechanisms (i.e., cytosine hypermethylation of CpG islands present in the promoter of these genes) may be responsible for this down-regulation. Protracted l-methionine (6.6 mmolkg for 15 days, twice a day) treatment in mice elicited in brain an increase of S-adenosyl-homocysteine, the processing product of the methyl donor S-adenosyl-methionine, and a marked decrease of reelin and GAD67 mRNAs in both WT and heterozygous reeler mice. This effect of l-methionine was associated with an increase in the number of methylated cytosines in the CpG island of the reelin promoter region. This effect was not observed for GAD65 or neuronal-specific enolase and was not replicated by glycine doses 2-fold greater than those of l-methionine. Prepulse inhibition of startle declined at a faster rate as the prepulsestartle interval increased in mice receiving l-methionine. Valproic acid (2 mmolkg for 15 days, twice a day) reverted l-methionine-induced down-regulation of reelin and GAD67 in both WT and heterozygous reeler mice, suggesting an epigenetic action through the inhibition of histone deacetylases. The same dose of valproate increased acetylation of histone H3 in mouse brain nearly 4-fold. This epigenetic mouse model may be useful in evaluating drug efficacy on schizophrenia vulnerability. Hence the inhibition of histone deacetylases could represent a pharmacological intervention mitigating epigenetically induced vulnerability to schizophrenia in individuals at risk.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Glutamate Decarboxylase/genetics , Isoenzymes/genetics , Schizophrenia/etiology , Acetylation , Animals , CpG Islands , DNA Methylation , Disease Susceptibility , Down-Regulation , Histones/metabolism , Methionine/pharmacology , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins , Promoter Regions, Genetic , RNA, Messenger/analysis , Reelin Protein , Reflex, Startle , Schizophrenia/genetics , Serine Endopeptidases , Valproic Acid/pharmacology
16.
Neuropharmacology ; 43(6): 925-37, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12423662

ABSTRACT

This review is designed to describe the evolution of the seminal observation made simultaneously in 1975 by Dr. W. Haefely's laboratory (Hoffman La Roche, Basel, Switzerland) and in the Laboratory of Preclinical Pharmacology (NIH, St. Elizabeths Hospital, Washington DC), that benzodiazepine action was mediated by a modulation of GABA action at GABA(A) receptors. In fact, our suggestion was that the benzodiazepine receptor was "a receptor on a receptor" and that this receptor was GABA(A). Needless to say, this suggestion created opposition, but we did not abandon the original idea, in fact, as shown in this review, there is now universal agreement with our hypothesis on the mode of action of benzodiazepines. Hence, this review deals with the allosteric modulation of GABA(A) receptors by benzodiazepines, the role of GABA(A) receptors and benzodiazepine structure diversities in this modulation, and describes the results of our attempts to establish a benzodiazepine (imidazenil) devoid of tolerance, withdrawal symptoms, and changes in the expression of GABA(A) receptor subunits during tolerance. It also deals with the idea that the synthesis of GABA(A) receptor subunits triggered by tolerance resides in dendrites and spines where mRNAs and the apparatus for this translation is located. New analytic procedures may foster progress in the understanding of tolerance to and withdrawal from benzodiazepines.


Subject(s)
Benzodiazepines/pharmacology , Dendrites/drug effects , Protein Subunits/physiology , Receptors, GABA-A/metabolism , Allosteric Regulation/physiology , Animals , Benzodiazepines/classification , Benzodiazepines/pharmacokinetics , Binding, Competitive , Chloride Channels/drug effects , Dendrites/metabolism , Diazepam/pharmacokinetics , Drug Tolerance/genetics , Drug Tolerance/physiology , GABA Modulators/pharmacokinetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , Imidazoles/pharmacokinetics , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Biological , Neocortex/cytology , Neocortex/metabolism , Pharmacokinetics , Protein Subunits/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/physiology , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substance Withdrawal Syndrome
17.
Mol Psychiatry ; 7(9): 1012-7, 2002.
Article in English | MEDLINE | ID: mdl-12399956

ABSTRACT

A polymorphic trinucleotide repeat (CGG/GCC) within the human Reelin gene (RELN) was examined as a candidate gene for autism spectrum disorders (ASDs). This gene encodes a large extracellular matrix protein that orchestrates neuronal positioning during corticogenesis. The CGG-repeat within the 5' untranslated region of RELN exon 1 was examined in 126 multiple-incidence families. The number of CGG repeats varied from three to 16 in affected individuals and controls, with no expansion or contraction observed during maternal (n = 291) or paternal (n = 287) transmissions in families with autistic probands. Although the frequencies of the RELN alleles and genotypes in affected children were not different from those in the comparison group, a family-based association test (FBAT) showed that the larger RELN alleles (> or = 11 repeats) were transmitted more often than expected to affected children (S = 43, E(S) = 34.5, P = 0.035); this was particularly the case for the 13-repeat RELN allele (S = 22, E(S) = 16, P = 0.034). Affected sib-pair (ASP) analysis found no evidence of excess sharing of RELN alleles in affected siblings. The impact of genotypes with large alleles (> or = 11 repeats) on the phenotypes in individuals with ASD was analyzed by ANOVA in a subset of the families for which results of the Autism Diagnostic Interview-Revised were available. Children with large RELN alleles did not show any difference in scores for questions related to the core symptoms of autistic disorder, but there was a tendency for children with at least one large RELN allele to have an earlier age at first phrase (chi(2) = 3.538, P = 0.06). Thus, although the case-control and affected sib-pair findings did not support a role for RELN in susceptibility to ASD, the more powerful family-based association study demonstrated that RELN alleles with larger numbers of CGG repeats may play a role in the etiology of some cases of ASD, especially in children without delayed phrase speech.


Subject(s)
Autistic Disorder/epidemiology , Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Case-Control Studies , Child, Preschool , Family Health , Female , Genetic Predisposition to Disease/epidemiology , Genotype , Humans , Infant , Male , Nerve Tissue Proteins , Phenotype , Reelin Protein , Serine Endopeptidases , Siblings , Trinucleotide Repeats/genetics
18.
Mol Interv ; 2(1): 47-57, 2002 Feb.
Article in English | MEDLINE | ID: mdl-14993361

ABSTRACT

The downregulation of the Reelin gene (RELN) that occurs in schizophrenic brains, which are characterized by pyramidal neurons with shortened dendrites and by reduced expression densities of dendritic spines, may well result from hypermethylation of the RELN promoter. In the adult mammalian brain, gamma-aminoburytic acid-secreting (GABAergic) interneurons release RELN into the extracellular matrix, where it binds with high affinity to the integrin receptors present at dendritic spine postsynaptic densities and likely plays a role, elaborated in this article, in synaptic plasticity. In heterozygous reeler mice, which are haploinsufficient in RELN, inhibitors of histone deacetylase increase DNA demethylase activity and restore RELN expression. Such inhibitors could thus be of therapeutic value in mitigating vulnerability to schizophrenia among high-risk individuals.


Subject(s)
Cell Adhesion Molecules, Neuronal/biosynthesis , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/biosynthesis , Histone Deacetylase Inhibitors , Nerve Tissue Proteins/biosynthesis , Schizophrenia/drug therapy , Schizophrenia/genetics , Serine Endopeptidases/biosynthesis , Animals , Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Humans , Nerve Tissue Proteins/physiology , Reelin Protein , Serine Endopeptidases/physiology
19.
Neurobiol Dis ; 8(5): 723-42, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11592844

ABSTRACT

In this review, we will first present a brief overview of the current understanding of: (a) the biology of reelin; (b) the putative reelin signaling pathways via integrin receptor stimulation; (c) the cytosolic adapter protein DAB1, which appears to be operative in the transduction of reelin's pleiotropic actions in embryonic, adolescent, and adult brain; (d) the regulation of GABAergic function, including some aspects of GABAergic system development; and (e) dendritic spine function and its role in the regulation of synaptic plasticity. We argue that a downregulation of reelin expression occurring in prefrontal cortex and in every brain structure of schizophrenia patients so far studied may be associated with a decrease in dendritic spine expression that in turn may provide an important reduction of cortical function as documented by the downregulation of glutamic acid decarboxylase67 (GAD67) expression, which might be secondary to a reduction of GABAergic axon terminals. This hypothesis is supported by a genetic mouse model of reelin haploinsufficiency that replicates the above-described dendritic and presynaptic GABAergic defects documented in schizophrenia brains.


Subject(s)
Brain/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Dendrites/ultrastructure , Extracellular Matrix Proteins/physiology , Nerve Tissue Proteins/physiology , Schizophrenia/etiology , gamma-Aminobutyric Acid/physiology , Adolescent , Adult , Age of Onset , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Bipolar Disorder/etiology , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Brain/embryology , Brain/growth & development , Brain/ultrastructure , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Count , Cell Movement , Child , Disease Models, Animal , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Fungal Proteins/genetics , Fungal Proteins/physiology , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Glutamate Decarboxylase/biosynthesis , Glutamate Decarboxylase/genetics , Heterozygote , Humans , Integrin alpha3 , Integrins/deficiency , Integrins/genetics , Integrins/physiology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Mental Disorders/metabolism , Mice , Mice, Knockout , Mice, Neurologic Mutants , Models, Neurological , Morphogenesis , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuronal Plasticity , Neurons/classification
20.
Arch Gen Psychiatry ; 57(11): 1061-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11074872

ABSTRACT

BACKGROUND: Reelin (RELN) is a glycoprotein secreted preferentially by cortical gamma-aminobutyric acid-ergic (GABAergic) interneurons (layers I and II) that binds to integrin receptors located on dendritic spines of pyramidal neurons or on GABAergic interneurons of layers III through V expressing the disabled-1 gene product (DAB1), a cytosolic adaptor protein that mediates RELN action. To replicate earlier findings that RELN and glutamic acid decarboxylase (GAD)(67), but not DAB1 expression, are down-regulated in schizophrenic brains, and to verify whether other psychiatric disorders express similar deficits, we analyzed, blind, an entirely new cohort of 60 postmortem brains, including equal numbers of patients matched for schizophrenia, unipolar depression, and bipolar disorder with nonpsychiatric subjects. METHODS: Reelin, GAD(65), GAD(67), DAB1, and neuron-specific-enolase messenger RNAs (mRNAs) and respective proteins were measured with quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) or Western blot analyses. Reelin-positive neurons were identified by immunohistochemistry using a monoclonal antibody. RESULTS: Prefrontal cortex and cerebellar expression of RELN mRNA, GAD(67) protein and mRNA, and prefrontal cortex RELN-positive cells was significantly decreased by 30% to 50% in patients with schizophrenia or bipolar disorder with psychosis, but not in those with unipolar depression without psychosis when compared with nonpsychiatric subjects. Group differences were absent for DAB1,GAD(65) and neuron-specific-enolase expression implying that RELN and GAD(67) down-regulations were unrelated to neuronal damage. Reelin and GAD(67) were also unrelated to postmortem intervals, dose, duration, or presence of antipsychotic medication. CONCLUSIONS: The selective down-regulation of RELN and GAD(67) in prefrontal cortex of patients with schizophrenia and bipolar disorder who have psychosis is consistent with the hypothesis that these parameters are vulnerability factors in psychosis; this plus the loss of the correlation between these 2 parameters that exists in nonpsychotic subjects support the hypothesis that these changes may be liability factors underlying psychosis.


Subject(s)
Bipolar Disorder/diagnosis , Bipolar Disorder/metabolism , Brain Chemistry , Brain/metabolism , Cell Adhesion Molecules, Neuronal/analysis , Extracellular Matrix Proteins/analysis , Phosphoproteins/analysis , Schizophrenia/diagnosis , Schizophrenia/metabolism , Schizosaccharomyces pombe Proteins , Activating Transcription Factor 1 , Adult , Blotting, Western , Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum/chemistry , Cerebellum/metabolism , Down-Regulation , Extracellular Matrix Proteins/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Nerve Tissue Proteins , Phosphoproteins/metabolism , Phosphopyruvate Hydratase/analysis , Phosphopyruvate Hydratase/metabolism , Prefrontal Cortex/chemistry , Prefrontal Cortex/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases
SELECTION OF CITATIONS
SEARCH DETAIL
...