Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34330827

ABSTRACT

There are no validated biomarkers for schizophrenia (SCZ), a disorder linked to neural network dysfunction. We demonstrate that collapsin response mediator protein-2 (CRMP2), a master regulator of cytoskeleton and, hence, neural circuitry, may form the basis for a biomarker because its activity is uniquely imbalanced in SCZ patients. CRMP2's activity depends upon its phosphorylation state. While an equilibrium between inactive (phosphorylated) and active (nonphosphorylated) CRMP2 is present in unaffected individuals, we show that SCZ patients are characterized by excess active CRMP2. We examined CRMP2 levels first in postmortem brains (correlated with neuronal morphometrics) and then, because CRMP2 is expressed in lymphocytes as well, in the peripheral blood of SCZ patients versus age-matched unaffected controls. In the brains and, more starkly, in the lymphocytes of SCZ patients <40 y old, we observed that nonphosphorylated CRMP2 was higher than in controls, while phosphorylated CRMP2 remained unchanged from control. In the brain, these changes were associated with dendritic structural abnormalities. The abundance of active CRMP2 with insufficient opposing inactive p-CRMP2 yielded a unique lowering of the p-CRMP2:CRMP2 ratio in SCZ patients, implying a disruption in the normal equilibrium between active and inactive CRMP2. These clinical data suggest that measuring CRMP2 and p-CRMP2 in peripheral blood might reflect intracerebral processes and suggest a rapid, minimally invasive, sensitive, and specific adjunctive diagnostic aid for early SCZ: increased CRMP2 or a decreased p-CRMP2:CRMP2 ratio may help cinch the diagnosis in a newly presenting young patient suspected of SCZ (versus such mimics as mania in bipolar disorder, where the ratio is high).


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Schizophrenia/diagnosis , Biomarkers/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Humans , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics
2.
Hum Mol Genet ; 27(2): 254-265, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29106556

ABSTRACT

Recent studies describe distinct DNA methylomes among phenotypic subclasses of neurons in the human brain, but variation in DNA methylation between common neuronal phenotypes distinguished by their function within distinct neural circuits remains an unexplored concept. Studies able to resolve epigenetic profiles at the level of microcircuits are needed to illuminate chromatin dynamics in the regulation of specific neuronal populations and circuits mediating normal and abnormal behaviors. The Illumina HumanMethylation450 BeadChip was used to assess genome-wide DNA methylation in stratum oriens GABAergic interneurons sampled by laser-microdissection from two discrete microcircuits along the trisynaptic pathway in postmortem human hippocampus from eight control, eight schizophrenia, and eight bipolar disorder subjects. Data were analysed using the minfi Bioconductor package in R software version 3.3.2. We identified 11 highly significant differentially methylated regions associated with a group of genes with high construct-validity, including multiple zinc finger of the cerebellum gene family members and WNT signaling factors. Genomic locations of differentially methylated regions were highly similar between diagnostic categories, with a greater number of differentially methylated individual cytosine residues between circuit locations in bipolar disorder cases than in schizophrenia or control (42, 7, and 7 differentially methylated positions, respectively). These findings identify distinct DNA methylomes among phenotypically similar populations of GABAergic interneurons functioning within separate hippocampal subfields. These data compliment recent studies describing diverse epigenotypes among separate neuronal subclasses, extending this concept to distinct epigenotypes within similar neuronal phenotypes from separate microcircuits within the human brain.


Subject(s)
Bipolar Disorder/genetics , GABAergic Neurons/physiology , Schizophrenia/genetics , Aged , Aged, 80 and over , Bipolar Disorder/physiopathology , Brain/metabolism , Cerebellum/metabolism , CpG Islands , DNA Methylation , Epigenesis, Genetic/genetics , Female , Genome , Hippocampus , Humans , Interneurons/metabolism , Male , Middle Aged , Neurons/metabolism , Schizophrenia/physiopathology , Signal Transduction , Temporal Lobe/metabolism
3.
Cereb Cortex ; 27(11): 5284-5293, 2017 11 01.
Article in English | MEDLINE | ID: mdl-27733539

ABSTRACT

GABAergic dysfunction in hippocampus, a key feature of schizophrenia (SZ), may contribute to cognitive impairment in this disorder. In stratum oriens (SO) of sector CA3/2 of the human hippocampus, a network of genes involved in the regulation of glutamic acid decarboxylase GAD67 has been identified. Several of the genes in this network including epigenetic factors histone deacetylase 1 (HDAC1) and death-associated protein 6 (DAXX), the GABAergic enzyme GAD65 as well as the kainate receptor (KAR) subunits GluR6 and 7 show significant changes in expression in this area in SZ. We have tested whether HDAC1 and DAXX regulate GAD67, GAD65, or GluR in the intact rodent hippocampus. Stereotaxic injections of lentiviral vectors bearing shRNAi sequences for HDAC1 and DAXX were delivered into the SO of CA3/2, followed by laser microdissection of individual transduced GABA neurons. Quantitative PCR (QPCR) analyses demonstrated that inhibition of HDAC1 and DAXX increased expression of GAD67, GAD65, and GluR6 mRNA. Inhibition of DAXX, but not HDAC1 resulted in a significant increase in GluR7 mRNA. Our data support the hypothesis that HDAC1 and DAXX play a central role in coordinating the expression of genes in the GAD67 regulatory pathway in the SO of CA3/2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CA2 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Epigenesis, Genetic , Glutamate Decarboxylase/metabolism , Histone Deacetylase 1/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , CA2 Region, Hippocampal/cytology , CA3 Region, Hippocampal/cytology , Cell Line , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Male , Molecular Chaperones , Neural Pathways/cytology , Neural Pathways/metabolism , Nuclear Proteins/antagonists & inhibitors , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism
4.
J Neurochem ; 138(1): 53-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27062510

ABSTRACT

The RNA integrity number (RIN) is often considered to be a critical measure of the quality of postmortem human brains. However, it has been suggested that RINs do not necessarily reflect the availability of intact mRNA. Using the Agilent bioanalyzer and qRT-PCR, we explored whether RINs provide a meaningful way of assessing mRNA degradation and integrity in human brain samples by evaluating the expression of 3'-5' mRNA sequences of the cytochrome C-1 (CYC1) gene. Analysis of electropherograms showed that RINs were not consistently correlated with RNA or cDNA profiles and appeared to be poor predictors of overall cDNA quality. Cycle thresholds from qRT-PCR analysis to quantify the amount of CYC1 mRNA revealed positive correlations of RINs with amplification of full-length transcripts, despite the variable degree of linear degradation along the 3'-5' sequence. These data demonstrate that in postmortem human brain tissue the RIN is an indicator of mRNA quantity independent of degradation, but does not predict mRNA integrity, suggesting that RINs provide an incomplete measure of brain tissue quality. Quality assessment of postmortem human brains by RNA integrity numbers (RINs) may be misleading, as they do not measure intact mRNAs. We show that the RIN is an indicator of mRNA quantity independent of degradation, but does not predict mRNA integrity, suggesting that RINs provide an incomplete measure of brain tissue quality. Our results resolve controversial assumption on interpreting quality assessments of human postmortem brains by RINs.


Subject(s)
Brain/metabolism , Brain/pathology , Cytochromes c1/genetics , RNA/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cells, Cultured , Fibroblasts , Gene Expression Profiling , Humans , Mental Disorders/pathology , Middle Aged , Neurodegenerative Diseases/pathology , Postmortem Changes , Predictive Value of Tests , RNA Stability/physiology , RNA, Messenger/metabolism , Young Adult
5.
Genesis ; 51(4): 234-45, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23349049

ABSTRACT

Postmortem studies have revealed a downregulation of the transcription factor Pax5 in GABAergic neurons in bipolar disorder, a neurodevelopmental disorder, raising the question whether Pax5 in GABAergic neurons has a role in normal brain development. In a genetic approach to study functions of Pax5 in GABAergic neurons, Pax5 was specifically deleted in GABAergic neurons from Pax5 floxed mice using a novel Gad1-Cre transgenic mouse line expressing Cre recombinase in Gad1-positive, that is, GABAergic neurons. Surprisingly, these mice developed a marked enlargement of the lateral ventricles at approximately 7 weeks of age, which was lethal within 1-2 weeks of its appearance. This hydrocephalus phenotype was observed in mice homozygous or heterozygous for the Pax5 conditional knockout, with a gene dosage-dependent penetrance. By QTL (quantitative trait loci) mapping, a 3.5 Mb segment on mouse chromosome 4 flanked by markers D4Mit237 and D4Mit214 containing approximately 92 genes including Pax5 has previously been linked to differences in lateral ventricular size. Our findings are consistent with Pax5 being a relevant gene underlying this QTL phenotype and demonstrate that Pax5 in GABAergic neurons is essential for normal ventricular development.


Subject(s)
Cerebral Ventricles/abnormalities , GABAergic Neurons/metabolism , Hydrocephalus/genetics , PAX5 Transcription Factor/genetics , Animals , Cerebral Ventricles/embryology , Chromosomes/genetics , Gene Dosage , Genetic Markers , Heterozygote , Homozygote , Mice , Mice, Transgenic , PAX5 Transcription Factor/metabolism , Penetrance , Phenotype , Physical Chromosome Mapping , Quantitative Trait Loci
6.
Handb Exp Pharmacol ; (213): 401-17, 2012.
Article in English | MEDLINE | ID: mdl-23027422

ABSTRACT

Studies of the hippocampus in postmortem brains from patients with schizophrenia and bipolar disorder have provided evidence for a defect of GABAergic interneurons. Significant decreases in the expression of GAD67, a marker for GABA cell function, have been found repeatedly in several different brain regions that include the hippocampus. In this region, nicotinic receptors are thought to play an important role in modulating the activity of GABAergic interneurons by influences of excitatory cholinergic afferents on their activity. In bipolar disorder, this influence appears to be particularly prominent in the stratum oriens of sectors CA3/2 and CA1, two sites where these cells constitute the exclusive neuronal cell type. In sector CA3/2, this layer receives a robust excitatory projection from the basolateral amygdala (BLA) and this is thought to play a central role in regulating GABA cells at this locus. Using laser microdissection, recent studies have focused selectively on these two layers and their associated GABA cells using microarray technology. The results have provided support for the idea that nicotinic cholinergic receptors play a particularly important role in regulating the activity of GABA neurons at these loci by regulating the progression of cell cycle and the repair of damaged DNA. In bipolar disorder, there is a prominent reduction in the expression of mRNAs for several different nicotinic subunit isoforms. These decreases could reflect a diminished influence of this receptor system on these GABA cells, particularly in sector CA3/2 where a preponderance of abnormalities have been observed in postmortem studies. In patients with bipolar disorder, excitatory nicotinic cholinergic fibers from the medial septum may converge with glutamatergic fibers from the BLA on GABAergic interneurons in the stratum oriens of CA3/2 and result in disturbances of their genomic and functional integrity, ones that may induce disruptions of the integration of microcircuitry within this region.


Subject(s)
Bipolar Disorder/physiopathology , Hippocampus/physiology , Interneurons/physiology , Receptors, Nicotinic/physiology , Schizophrenia/physiopathology , gamma-Aminobutyric Acid/physiology , Glutamate Decarboxylase/physiology , Humans , Neurons, Afferent/physiology
7.
Proc Natl Acad Sci U S A ; 106(28): 11731-6, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19564623

ABSTRACT

GABA cell dysfunction in both schizophrenia (SZ) and bipolar disorder (BD) involves decreased GAD(67) expression, although this change involves fundamentally different networks of genes in the 2 disorders. One gene that is common to these 2 networks is cyclin D2, a key component of cell cycle regulation that shows increased expression in SZ, but decreased expression in BD. Because of the importance of cell cycle regulation in maintaining functional differentiation and DNA repair, the current study has examined the genes involved in the G(1) and G(2) checkpoints to generate new hypotheses regarding the regulation of the GABA cell phenotype in the hippocampus of SZ and BD. The results have demonstrated significant changes in cell cycle regulation in both SZ and BD and these changes include the transcriptional complex (TC) that controls the expression of E2F/DP-1 target genes critical for progression to G(2)/M. The methyl-CpG binding domain protein (MBD4) that is pivotal for DNA repair, is significantly up-regulated in the stratum oriens (SO) of CA3/2 and CA1 in SZs and BDs. However, other genes associated with the TC, and the G(1) and G(2) checkpoints, show complex changes in expression in the SO of CA3/2 and CA1 of both SZs and BDS. Overall, the patterns of expression observed have suggested that the regulation of functional differentiation and/or genomic integrity of hippocampal GABA cells varies according to diagnosis and their location within the trisynaptic pathway.


Subject(s)
Bipolar Disorder/metabolism , Cell Cycle/physiology , Hippocampus/cytology , Neurons/metabolism , Schizophrenia/metabolism , DNA Repair/physiology , Endodeoxyribonucleases/metabolism , Gene Expression Regulation/physiology , Glutamate Decarboxylase/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , gamma-Aminobutyric Acid/metabolism
8.
Proc Natl Acad Sci U S A ; 105(52): 20935-40, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19104056

ABSTRACT

Significant reductions in GABAergic cell numbers and/or activity have been demonstrated in the hippocampus of subjects with schizophrenia and bipolar disorder. To understand how different subpopulations of interneurons are regulated, laser microdissection and gene expression profiling have been used to "deconstruct" the trisynaptic pathway, so that subtypes of GABA cells could be defined by their location in various layers of CA3/2 and CA1. The results suggest that the cellular endophenotypes for SZ and BD may be determined by multiple factors that include unique susceptibility genes for the respective disorders and altered integration among hippocampal GABA cells with extrinsic and intrinsic afferent fiber systems. The extensive and intricate data that has come from this study has provided insights into how a complex circuit, like the trisynaptic pathway, may be regulated in human hippocampus in both health and disease.


Subject(s)
Bipolar Disorder/metabolism , Gene Expression Profiling , Gene Expression Regulation , Hippocampus/metabolism , Schizophrenia/metabolism , Synapses/metabolism , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Female , Hippocampus/pathology , Humans , Male , Microdissection , Schizophrenia/genetics , Schizophrenia/pathology , Synapses/pathology , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
9.
Trends Neurosci ; 31(5): 234-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18395805

ABSTRACT

Many risk genes interact synergistically to produce schizophrenia and many neurotransmitter interactions have been implicated. We have developed a circuit-based framework for understanding gene and neurotransmitter interactions. NMDAR hypofunction has been implicated in schizophrenia because NMDAR antagonists reproduce symptoms of the disease. One action of antagonists is to reduce the excitation of fast-spiking interneurons, resulting in disinhibition of pyramidal cells. Overactive pyramidal cells, notably those in the hippocampus, can drive a hyperdopaminergic state that produces psychosis. Additional aspects of interneuron function can be understood in this framework, as follows. (i) In animal models, NMDAR antagonists reduce parvalbumin and GAD67, as found in schizophrenia. These changes produce further disinhibition and can be viewed as the aberrant response of a homeostatic system having a faulty activity sensor (the NMDAR). (ii) Disinhibition decreases the power of gamma oscillation and might thereby produce negative and cognitive symptoms. (iii) Nicotine enhances the output of interneurons, and might thereby contribute to its therapeutic effect in schizophrenia.


Subject(s)
Nerve Net , Neurotransmitter Agents/genetics , Receptors, N-Methyl-D-Aspartate , Schizophrenia/genetics , Animals , Cognition/physiology , Dopamine/metabolism , Gene Expression , Hippocampus/cytology , Hippocampus/metabolism , Homeostasis , Humans , N-Methylaspartate/metabolism , Neurotransmitter Agents/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Risk , gamma-Aminobutyric Acid/metabolism
10.
Brain ; 132(Pt 7): 1795-809, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19052140

ABSTRACT

Parkinson's disease is caused by a progressive loss of the midbrain dopamine (DA) neurons in the substantia nigra pars compacta. Although the main cause of Parkinson's disease remains unknown, there is increasing evidence that it is a complex disorder caused by a combination of genetic and environmental factors, which affect key signalling pathways in substantia nigra DA neurons. Insights into pathogenesis of Parkinson's disease stem from in vitro and in vivo models and from postmortem analyses. Recent technological developments have added a new dimension to this research by determining gene expression profiles using high throughput microarray assays. However, many of the studies reported to date were based on whole midbrain dissections, which included cells other than DA neurons. Here, we have used laser microdissection to isolate single DA neurons from the substantia nigra pars compacta of controls and subjects with idiopathic Parkinson's disease matched for age and postmortem interval followed by microarrays to analyse gene expression profiling. Our data confirm a dysregulation of several functional groups of genes involved in the Parkinson's disease pathogenesis. In particular, we found prominent down-regulation of members of the PARK gene family and dysregulation of multiple genes associated with programmed cell death and survival. In addition, genes for neurotransmitter and ion channel receptors were also deregulated, supporting the view that alterations in electrical activity might influence DA neuron function. Our data provide a 'molecular fingerprint identity' of late-stage Parkinson's disease DA neurons that will advance our understanding of the molecular pathology of this disease.


Subject(s)
Dopamine/metabolism , Gene Expression Profiling/methods , Neurons/metabolism , Parkinson Disease/genetics , Substantia Nigra/metabolism , Aged , Aged, 80 and over , Apoptosis/genetics , Cell Survival/genetics , Cytoskeleton/pathology , Dopamine/genetics , Female , Gene Expression Regulation , Humans , Ion Channels/genetics , Ion Channels/metabolism , Male , Microdissection/methods , Mitochondria/physiology , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phenotype , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Substantia Nigra/pathology , Synapses/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Cereb Cortex ; 18(7): 1529-35, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17971342

ABSTRACT

Previous work in animal models has shown that projections from the basolateral amygdala (BLA) progressively infiltrate the medial prefrontal cortex (mPFC) from birth to adulthood, with the most dramatic sprouting occurring during the postweanling period. GABAergic (gamma-aminobutyric acidergic) interneurons in the human homolog of the rat mPFC have been implicated in the pathophysiology of schizophrenia, an illness with an onset that is delayed until late adolescence. Here we investigated the interaction of BLA fibers with mPFC GABAergic interneurons from postnatal day 6 (P6) to P120 using anterograde tracing and immunocytochemistry. We found a 3-fold increase in axosomatic and an 8-fold increase in axo-dendritic contacts in both layers II and V of the mPFC. Ultrastructural analysis using a colloidal gold immunolocalization demonstrated that the greatest proportion of BLA appositions were with GABA-negative spines (30.8%) and GABA-positive dendritic shafts (35.5%). Although GABA-negative interactions demonstrated well-defined axo-spinous synapses, membrane specializations could not be identified with confidence in GABA-positive elements. Our findings suggest that GABAergic interneurons are major targets for BLA fibers projecting to the mPFC. The establishment of this circuitry, largely during adolescence, may contribute to the integration of emotional responses with attentional and other cognitive processes mediated within this region during corticolimbic development.


Subject(s)
Aging/physiology , Amygdala/physiology , Efferent Pathways/physiology , Interneurons/physiology , Nerve Net/physiology , Prefrontal Cortex/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
12.
Neuron ; 34(2): 289-300, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11970870

ABSTRACT

Auditory information critical for fear conditioning, a model of emotional learning, is conveyed to the lateral nucleus of the amygdala via two routes: directly from the medial geniculate nucleus and indirectly from the auditory cortex. Here we show in the cortico-amygdala pathway that learned fear occludes electrically induced long-term potentiation (LTP). Quantal analysis of the expression of LTP in this pathway reveals a significant presynaptic component reflected in an increase in probability of transmitter release. Conditioned fear also is accompanied by the enhancement in transmitter release at this cortico-amygdala synapse. These results indicate that the synaptic projections from the auditory cortex to the lateral amygdala are modified during the acquisition and expression of fear to auditory stimulation, thus further strengthening the proposed link between LTP in the auditory pathways to the amygdala and learned fear.


Subject(s)
Amygdala/physiology , Cerebral Cortex/physiology , Conditioning, Psychological/physiology , Fear/physiology , Long-Term Potentiation/physiology , Presynaptic Terminals/physiology , Synaptic Transmission/physiology , Animals , Electrophysiology , In Vitro Techniques , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Synapses/physiology
13.
Schizophr Res ; 201: 270-277, 2018 11.
Article in English | MEDLINE | ID: mdl-29759351

ABSTRACT

Deep layer III pyramidal cells in the dorsolateral prefrontal cortex (DLPFC) from subjects with schizophrenia and bipolar disorder previously were shown to exhibit dendritic arbor pathology. This study sought to determine whether MARCKS, its regulatory protein dysbindin-1, and two proteins, identified using microarray data, CDC42BPA and ARHGEF6, were associated with dendritic arbor pathology in the DLPFC from schizophrenia and bipolar disorder subjects. Using western blotting, relative protein expression was assessed in the DLPFC (BA 46) grey matter from subjects with schizophrenia (n = 19), bipolar disorder (n = 17) and unaffected control subjects (n = 19). Protein expression data were then correlated with dendritic parameter data obtained previously. MARCKS and dysbindin-1a expression levels did not differ among the three groups. Dysbindin-1b expression was 26% higher in schizophrenia subjects (p = 0.01) and correlated inversely with basilar dendrite length (r = -0.31, p = 0.048) and the number of spines per basilar dendrite (r = -0.31, p = 0.048), but not with dendritic spine density (r = -0.16, p = 0.32). The protein expression of CDC42BPA was 33% higher in schizophrenia subjects (p = 0.03) but, did not correlate with any dendritic parameter (p > 0.05). ARHGEF6 87 kDa isoform expression did not differ among the groups. CDC42BPA expression was not altered in frontal cortex from rats chronically administered haloperidol or clozapine. Dysbindin-1b appears to play a role in dendritic arbor pathology observed previously in the DLPFC in schizophrenia.


Subject(s)
Dendrites/metabolism , Dysbindin/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Animals , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Cohort Studies , Dendrites/drug effects , Dendrites/pathology , Disease Models, Animal , Female , Gene Expression/drug effects , Gray Matter/drug effects , Gray Matter/metabolism , Gray Matter/pathology , Humans , Male , Middle Aged , Myotonin-Protein Kinase/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , Psychotropic Drugs/pharmacology , Psychotropic Drugs/therapeutic use , Rats , Rho Guanine Nucleotide Exchange Factors/metabolism , Schizophrenia/drug therapy , Schizophrenia/pathology
14.
Schizophr Res ; 93(1-3): 33-41, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17442540

ABSTRACT

UNLABELLED: Apoptosis is thought to contribute to neuronal loss in bipolar disorder and schizophrenia, although empiric evidence in support of this idea has been lacking. In this study, we investigated whether or not apoptosis is associated with GABAergic interneurons in the anterior cingulate cortex in schizophrenia (n=14) and bipolar disorder (n=14) when compared to normal controls (n=14). A double-labeling technique using the Klenow method of in situ end-labeling (ISEL) of single-stranded DNA breaks was combined with an in situ hybridization localization of mRNA for the 67 kDa isoform of glutamate decarboxylase (GAD67) and applied to the anterior cingulate cortex of 14 normal controls, 14 schizophrenics, and 14 patients with bipolar disorder matched for age and postmortem interval. An increase in Klenow-positive, GAD67-negative nuclei were observed in layer V/VI of patients with bipolar disorder, but not schizophrenics. Klenow-positive cells that were also positive for GAD67 mRNA did not show differences in either patient group. CONCLUSIONS: This is the first demonstration that there is more DNA fragmentation in cells showing no detectable GAD67 mRNA in patients with bipolar disorder than in schizophrenics or controls. These findings suggest that non-GABAergic cells may be selectively vulnerable to oxidative stress in patients with bipolar disorder.


Subject(s)
Apoptosis/genetics , Bipolar Disorder/genetics , DNA Fragmentation , Glutamate Decarboxylase/genetics , Gyrus Cinguli/pathology , Isoenzymes/genetics , Schizophrenia/genetics , gamma-Aminobutyric Acid/physiology , Adult , Aged , Aged, 80 and over , Bipolar Disorder/pathology , Cell Count , Cohort Studies , Dominance, Cerebral/physiology , Female , Humans , Interneurons/pathology , Male , Middle Aged , Oxidative Stress/physiology , RNA, Messenger/genetics , Schizophrenia/pathology
15.
Schizophr Res ; 96(1-3): 46-61, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17698324

ABSTRACT

The aim of this study was to examine whether glutamatergic inputs onto GABA interneurons via the kainate receptor in the anterior cingulate cortex may be altered in schizophrenia and bipolar disorder. Hence, in a cohort of 60 post-mortem human brains from schizophrenia, bipolar disorder, and normal control subjects, we simultaneously labeled the mRNA for the GluR5 or GluR6 subunit of the kainate receptor with [(35)S] and the mRNA for the 67 kD isoform of the GABA synthesizing enzyme glutamic acid decarboxylase (GAD)(67) with digoxigenin using an immunoperoxidase method. The density of the GAD(67) mRNA-containing neurons that co-expressed GluR5 mRNA was decreased by 43% and 40% in layer 2 of the anterior cingulate cortex in schizophrenia and bipolar disorder, respectively. In contrast, the density of the GAD(67) mRNA-containing cells that expressed GluR6 mRNA was unaltered in either condition. Furthermore, the amount of GluR5 or GluR6 mRNA in the GAD(67) mRNA-expressing cells that contained a detectable level of these transcripts was also unchanged. Finally, the density of cells that did not contain GAD(67) mRNA, which presumably included all pyramidal neurons, but expressed the mRNA for the GluR5 or GluR6 subunit was not altered. Thus, glutamatergic modulation of inhibitory interneurons, but not pyramidal neurons, via kainate receptors containing the GluR5 subunit appears to be selectively altered in the anterior cingulate cortex in schizophrenia and bipolar disorder.


Subject(s)
Bipolar Disorder/metabolism , Gyrus Cinguli/physiopathology , Receptors, Kainic Acid/metabolism , Schizophrenia/metabolism , Adolescent , Adult , Antipsychotic Agents/therapeutic use , Biopsy , Bipolar Disorder/drug therapy , Bipolar Disorder/pathology , Child , Female , Functional Laterality , Gyrus Cinguli/anatomy & histology , Gyrus Cinguli/pathology , Humans , Male , Protein Subunits/metabolism , Schizophrenia/drug therapy , Schizophrenia/pathology
16.
Schizophr Bull ; 33(4): 932-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17575303

ABSTRACT

The endophenotype is a construct that has utility for the study of postmortem brains from patients with psychotic disorders. By identifying networks of genes that show changes in expression within specific neuronal populations implicated in the pathophysiology of schizophrenia and bipolar disorder, it may be possible to move toward understanding these disorders at the cellular and molecular levels. The ultimate goal is to characterize their respective underlying genotypes.


Subject(s)
Bipolar Disorder , Nerve Net , Phenotype , Schizophrenia , Amygdala/metabolism , Amygdala/pathology , Amygdala/physiopathology , Antioxidants/physiology , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Calcium Channels, L-Type/physiology , Down-Regulation , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Nerve Net/metabolism , Nerve Net/pathology , Nerve Net/physiopathology , Schizophrenia/genetics , Schizophrenia/pathology , Schizophrenia/physiopathology , gamma-Aminobutyric Acid/physiology
17.
Genes (Basel) ; 8(5)2017 May 15.
Article in English | MEDLINE | ID: mdl-28505127

ABSTRACT

Identification of 108 genomic regions significantly associated with schizophrenia risk by the Psychiatric Genomics Consortium was a milestone for the field, and much work is now focused on determining the mechanism of risk associated with each locus. Within these regions, we investigated variability of DNA methylation, a low-level cellular phenotype closely linked to genotype, in two highly similar cellular populations sampled from the human hippocampus, to draw inferences about the elaboration of genotype to phenotype within these loci enriched for schizophrenia risk. DNA methylation was assessed with the Illumina HumanMethylation450 BeadArray in tissue laser-microdissected from the stratum oriens of subfield CA1 or CA2/3, regions having unique connectivity with intrinsic and extrinsic fiber systems within the hippocampus. Samples consisted of postmortem human hippocampus tissue from eight schizophrenia patients, eight bipolar disorder patients, and eight healthy control subjects. Within these genomic regions, we observed far greater difference in methylation patterns between circuit locations within subjects than in a single subregion between subjects across diagnostic groups, demonstrating the complexity of genotype to phenotype elaboration across the diverse circuitry of the human brain.

18.
Schizophr Res ; 73(1): 79-89, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15567080

ABSTRACT

Using a two-dimensional cell counting approach, a 1991 study in the anterior cingulate cortex (ACCx) detected a reduction in the density of nonpyramidal neurons in layers II-VI of schizophrenic subjects. Schizophrenics without superimposed mood disturbances showed a 16% decrease in layer II, while schizoaffectives showed a 30% decrease, suggesting that a decreased density of nonpyramidal neurons in layer II of ACCx might vary more strongly with affective disorder than with schizophrenia. Two follow-up studies from this laboratory, one a replication of that reported in 1991 and the other an analysis of tyrosine hydroxylase immunoreactive fibers, were undertaken in ACCx of normal controls and schizophrenics. These three data sets have been combined and a meta-analysis of the density of pyramidal, nonpyramidal and glial cells was performed to explore whether changes in the density of interneurons in ACCx may be a reliable finding in the major psychoses. Not all groups have reported this finding, but several had employed a different cell counting technique (i.e. three dimensional optical dissector), which could help to explain the discrepant findings in schizophrenia and affective disorder. The data from each of three different studies (now designated as studies A, B and C, respectively) have been internally normalized, combined into a single dataset and analyzed using nonparametric statistics. Tissue blocks from a subset of cases in study B (six controls, six schizophrenics and six bipolars) were embedded in celloidin and counted using an "unbiased" three dimensional counting method (study D). The data from studies A and B indicate that the density of nonpyramidal neurons in layer II of ACCx in the schizoaffective and bipolar samples was significantly decreased. In the schizophrenics, the nonpyramidal neurons were also decreased, but only by 15%. All three groups also showed a decrease of pyramidal neurons in layers IV, V and VI, but this difference was significant only in layer IV of the schizophrenics. When data from study C were added, the differences in pyramidal and nonpyramidal neurons were less striking. For study D, the pattern of findings are strikingly similar to those obtained in studies A, B and C, indicating that both 2D and 3D cell counting methodologies are capable of detecting the same differences. Taken together, these results indicate that the earlier finding of a decreased density of nonpyramidal neurons in ACCx of schizophrenics is consistent across non-overlapping subjects and/or methods in four separate studies, and is more pronounced in schizoaffective and bipolar subjects than in schizophrenics without superimposed mood disturbance.


Subject(s)
Bipolar Disorder/pathology , Gyrus Cinguli/pathology , Imaging, Three-Dimensional , Schizophrenia/pathology , Cell Count , Cohort Studies , Humans , Interneurons/pathology , Neuroglia/pathology , Pyramidal Cells/pathology
19.
Arch Gen Psychiatry ; 60(4): 359-64, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12695312

ABSTRACT

BACKGROUND: Apoptosis is thought to play a role in neuronal pathology in schizophrenia and bipolar disorder. METHODS: To test this hypothesis, the Klenow method for in situ end-labeling of single-stranded DNA breaks was applied to anterior cingulate cortex from 18 healthy controls, 18 schizophrenic subjects, and 10 bipolar subjects. RESULTS: An unexpected reduction (71%) in Klenow-positive nuclei was found in schizophrenic but not in bipolar cortexes. CONCLUSIONS: To our knowledge to date, this is the first demonstration that there is much less DNA fragmentation in individuals with schizophrenia than in healthy controls and bipolar subjects, which raises a key question as to whether this alteration represents an adaptive or nonadaptive change in the regulation of intracellular signaling and mitochondrial oxidative pathways associated with apoptosis.


Subject(s)
Apoptosis/genetics , Bipolar Disorder/genetics , DNA Fragmentation/genetics , Schizophrenia/genetics , Adolescent , Adult , Aged , Bipolar Disorder/metabolism , Bipolar Disorder/physiopathology , Cohort Studies , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Female , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiopathology , Humans , In Situ Nick-End Labeling , Male , Middle Aged , Polymorphism, Single-Stranded Conformational , Schizophrenia/metabolism , Schizophrenia/physiopathology
20.
Arch Gen Psychiatry ; 61(7): 649-57, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15237077

ABSTRACT

BACKGROUND: Disturbances of gamma-aminobutyric acid interneurons in the cerebral cortex contribute to the pathophysiology of schizophrenia and bipolar disorder. The activity of these neurons is, in turn, modulated by glutamatergic inputs furnished by pyramidal neurons. OBJECTIVE: To test the hypothesis that glutamatergic inputs onto gamma-aminobutyric acid interneurons via the N-methyl-d-aspartate (NMDA) receptor are altered in the anterior cingulate cortex in schizophrenia and bipolar disorder. DESIGN: A double in situ hybridization technique was used to simultaneously label the messenger RNA (mRNA) for the NMDA NR(2A) subunit with (35)sulfur and the mRNA for the 67-kDa isoform of the gamma-aminobutyric acid synthesizing enzyme glutamic acid decarboxylase (GAD(67)) with digoxigenin. SETTING: Postmortem human brain studies. PARTICIPANTS: We studied 17 subjects with schizophrenia, 17 subjects with bipolar disorder, and 17 normal control subjects. RESULTS: The density of all GAD(67) mRNA-containing neurons was decreased by 53% and 28%, in layers 2 and 5, respectively, in subjects with schizophrenia, whereas in subjects with bipolar disorder there was a 35% reduction in layer 2 only. For GAD(67) mRNA-containing neurons that co-expressed NR(2A)mRNA, their numerical density was decreased by 73% and 52%, in layers 2 and 5, respectively, in subjects with schizophrenia and by 60% in layer 2 in those with bipolar disorder. In the schizophrenia group, the density of the GAD(67)mRNA-containing neurons that did not co-express NR(2A)mRNA was also decreased by 42% in layer 2. In both disease groups, the expression level of NR(2A)mRNA in GAD(67) mRNA-containing cells was unaltered. CONCLUSIONS: The density of gamma-aminobutyric acid interneurons that express the NMDA NR(2A)subunit appears to be decreased in schizophrenia and bipolar disorder. Future studies will address whether subpopulations of these neurons may be differentially affected in the 2 conditions.


Subject(s)
Bipolar Disorder/metabolism , Glutamate Decarboxylase/metabolism , Gyrus Cinguli/metabolism , Interneurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/metabolism , gamma-Aminobutyric Acid/biosynthesis , Adult , Aged , Bipolar Disorder/enzymology , Cell Count , Digoxigenin/metabolism , Female , Gene Expression , Glutamate Decarboxylase/genetics , Gyrus Cinguli/cytology , Gyrus Cinguli/enzymology , Humans , In Situ Hybridization , Interneurons/cytology , Interneurons/enzymology , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Middle Aged , Pyramidal Cells/cytology , Pyramidal Cells/enzymology , Pyramidal Cells/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Schizophrenia/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL