Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Neurosci Res ; 92(10): 1307-18, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24801281

ABSTRACT

Neurotrophins, such as brain-derived neurotrophic factor (BDNF), are initially expressed in a precursor form (e.g., pro-BDNF) and cleaved to form mature BDNF (mBDNF). After pilocarpine-induced status epilepticus (SE), increases in neurotrophins regulate a wide variety of cell-signaling pathways, including prosurvival and cell-death machinery in a receptor-specific manner. Pro-BDNF preferentially binds to the p75 neurotrophin receptor (p75(NTR) ), whereas mBDNF is the major ligand of the tropomyosin-related kinase receptor. To elucidate a potential role for p75(NTR) in acute stages of epileptogenesis, rats were injected prior to and at onset of SE with LM11A-31, a small-molecule ligand that binds to p75(NTR) to promote survival signaling and inhibit neuronal cell death. Modulation of early p75(NTR) signaling and its effects on electrographic SE, SE-induced neurodegeneration, and subsequent spontaneous seizures were examined after LM11A-31 administration. Despite an established neuroprotective effect of LM11A-31 in several animal models of neurodegenerative disorders (e.g., Alzheimer's disease, traumatic brain injury, and spinal cord injury), high-dose LM11A-31 administration prior to and at onset of SE did not reduce the intensity of electrographic SE, prevent SE-induced neuronal cell injury, or inhibit the progression of epileptogenesis. Further studies are required to understand the role of p75(NTR) activation during epileptogenesis and in seizure-induced cell injury in the hippocampus, among other potential cellular pathologies contributing to the onset of spontaneous seizures. Additional studies utilizing more prolonged treatment with LM11A-31 are required to reach a definite conclusion on its potential neuroprotective role in epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Isoleucine/analogs & derivatives , Morpholines/therapeutic use , Receptors, Nerve Growth Factor/metabolism , Status Epilepticus/drug therapy , Analysis of Variance , Animals , Anticonvulsants/blood , Brain Waves/drug effects , Disease Models, Animal , Electroencephalography , Fluoresceins , Isoleucine/blood , Isoleucine/therapeutic use , Morpholines/blood , Muscarinic Agonists/toxicity , Nerve Tissue Proteins , Pilocarpine/toxicity , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Receptors, Nerve Growth Factor/chemistry , Spectrum Analysis , Status Epilepticus/chemically induced , Time Factors
2.
Neurobiol Dis ; 62: 73-85, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24051278

ABSTRACT

Pilocarpine-induced status epilepticus (SE), which results in temporal lobe epilepsy (TLE) in rodents, activates the JAK/STAT pathway. In the current study, we evaluate whether brief exposure to a selective inhibitor of the JAK/STAT pathway (WP1066) early after the onset of SE affects the severity of SE or reduces later spontaneous seizure frequency via inhibition of STAT3-regulated gene transcription. Rats that received systemic WP1066 or vehicle at the onset of SE were continuously video-EEG monitored during SE and for one month to assess seizure frequency over time. Protein and/or mRNA levels for pSTAT3, and STAT3-regulated genes including: ICER, Gabra1, c-myc, mcl-1, cyclin D1, and bcl-xl were evaluated in WP1066 and vehicle-treated rats during stages of epileptogenesis to determine the acute effects of WP1066 administration on SE and chronic epilepsy. WP1066 (two 50mg/kg doses) administered within the first hour after onset of SE results in transient inhibition of pSTAT3 and long-term reduction in spontaneous seizure frequency. WP1066 alters the severity of chronic epilepsy without affecting SE or cell death. Early WP1066 administration reduces known downstream targets of STAT3 transcription 24h after SE including cyclin D1 and mcl-1 levels, known for their roles in cell-cycle progression and cell survival, respectively. These findings uncover a potential effect of the JAK/STAT pathway after brain injury that is physiologically important and may provide a new therapeutic target that can be harnessed for the prevention of epilepsy development and/or progression.


Subject(s)
Brain/physiopathology , Pyridines/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Status Epilepticus/drug therapy , Tyrphostins/therapeutic use , Animals , Brain/drug effects , Cell Death , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Disease Models, Animal , Electroencephalography , Hippocampus/drug effects , Hippocampus/metabolism , Phosphorylation , Pilocarpine , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Seizures/drug therapy , Signal Transduction/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/physiopathology , Tyrphostins/pharmacokinetics
3.
Neuroscience ; 168(3): 797-810, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20381588

ABSTRACT

Disruption of the GABAergic system has been implicated in multiple developmental disorders, including epilepsy, autism spectrum disorder and schizophrenia. The human gene encoding uPAR (PLAUR) has been shown recently to be associated with the risk of autism. The uPAR(-/-) mouse exhibits a regionally-selective reduction in GABAergic interneurons in frontal and parietal regions of the cerebral cortex as well as in the CA1 and dentate gyrus subfields of the hippocampus. Behaviorally, these mice exhibit increased sensitivity to pharmacologically-induced seizures, heightened anxiety, and atypical social behavior. Here, we explore potential alterations in GABAergic circuitry that may occur in the context of altered interneuron development. Analysis of gene expression for 13 GABA(A) receptor subunits using quantitative real-time polymerase chain reaction (PCR) indicates seven subunit mRNAs (alpha(1), alpha(2), alpha(3), beta(2), beta(3), gamma(2S) and gamma(2L)) of interest. Semi-quantitative in situ hybridization analysis focusing on these subunit mRNAs reveals a complex pattern of potential gene regulatory adaptations. The levels of alpha(2) subunit mRNAs increase in frontal cortex, CA1 and CA3, while those of alpha3 decrease in frontal cortex and CA1. In contrast, alpha(1) subunit mRNAs are unaltered in any region examined. beta(2) subunit mRNAs are increased in frontal cortex whereas beta(3) subunit mRNAs are decreased in parietal cortex. Finally, gamma(2S) subunit mRNAs are increased in parietal cortex while gamma(2L) subunit mRNAs are increased in the dentate gyrus, potentially altering the gamma(2S):gamma(2L) ratio in these two regions. For all subunits, no changes were observed in forebrain regions where GABAergic interneuron numbers are normal. We propose that disrupted differentiation of GABAergic neurons specifically in frontal and parietal cortices leads to regionally-selective alterations in local circuitry and subsequent adaptive changes in receptor subunit composition. Future electrophysiological studies will be useful in determining how alterations in network activity in the cortex and hippocampus relate to the observed behavioral phenotype.


Subject(s)
Child Development Disorders, Pervasive/genetics , Receptors, GABA-A/biosynthesis , Receptors, Urokinase Plasminogen Activator/physiology , Telencephalon/metabolism , Animals , Child , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/biosynthesis , Protein Subunits/genetics , RNA, Messenger/biosynthesis , Receptors, GABA-A/genetics , Receptors, Urokinase Plasminogen Activator/genetics
4.
Proc Natl Acad Sci U S A ; 102(33): 11894-9, 2005 Aug 16.
Article in English | MEDLINE | ID: mdl-16091474

ABSTRACT

GABA is the major inhibitory transmitter at CNS synapses. Changes in subunit composition of the pentameric GABA(A) receptor, including increased levels of alpha4 subunit in dentate granule cells and associated functional alterations such as increased zinc blockade of GABA currents, are hypothesized to be critical components of epileptogenesis. Here, we report that the minimal promoter of the human alpha4 subunit gene (GABRA4p), when used to drive reporter gene expression from adeno-associated viral vectors, controls condition-specific up-regulation in response to status epilepticus, defining a transcriptional mechanism for seizure-induced changes in levels of alpha4 subunit containing GABA(A) receptors. Transfection studies in primary hippocampal neurons show that inducible early growth response factor 3 (Egr3) up-regulates GABRA4p activity as well as the levels of endogenous alpha4 subunits. Given that Egr3 knockout mice display approximately 50% less GABRA4 mRNAs in the hippocampus and that increases in alpha4 and Egr3 mRNAs in response to pilocarpine-induced status epilepticus are accompanied by increased binding of Egr3 to GABRA4 in dentate granule cells, our findings support a role for Egr3 as a major regulator of GABRA4 in developing neurons and in epilepsy.


Subject(s)
DNA-Binding Proteins/metabolism , Promoter Regions, Genetic/genetics , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Seizures/genetics , Seizures/metabolism , Transcription Factors/metabolism , Up-Regulation , Animals , Base Sequence , Brain/metabolism , Brain/pathology , Cells, Cultured , DNA-Binding Proteins/genetics , Dependovirus/genetics , Early Growth Response Protein 3 , Humans , Male , Molecular Sequence Data , Protein Kinase C/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Seizures/pathology , Sequence Alignment , Transcription Factors/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics
5.
Proc Natl Acad Sci U S A ; 102(20): 7380-5, 2005 May 17.
Article in English | MEDLINE | ID: mdl-15870187

ABSTRACT

Benzodiazepines remain widely used for the treatment of anxiety disorders despite prominent, often limiting side effects including sedation, muscle relaxation, and ataxia. A compound producing a robust anxiolytic action comparable to benzodiazepines, but lacking these limiting side effects at therapeutic doses (an anxioselective agent), would represent an important advance in the treatment of generalized anxiety disorder, and perhaps other anxiety disorders. Here we report that the pyrazolo[1,5-a]-pyrimidine, ocinaplon, exhibits an anxioselective profile in both preclinical procedures and in patients with generalized anxiety disorder, the most common of the anxiety disorders. In rats, ocinaplon produces significant muscle relaxation, ataxia, and sedation only at doses >25-fold higher than the minimum effective dose (3.1 mg/kg) in the Vogel "conflict" test. This anticonflict effect is blocked by flumazenil (Ro 15-1788), indicating that like benzodiazepines, ocinaplon produces an anxiolytic action through allosteric modulation of GABA(A) receptors. Nonetheless, in eight recombinant GABA(A) receptor isoforms expressed in Xenopus oocytes, the potency and efficacy of ocinaplon to potentiate GABA responses varied with subunit composition not only in an absolute sense, but also relative to the prototypical benzodiazepine, diazepam. In a double blind, placebo controlled clinical trial, a 2-week regimen of ocinaplon (total daily dose of 180-240 mg) produced statistically significant reductions in the Hamilton rating scale for anxiety scores. In this study, the incidence of benzodiazepine-like side effects (e.g., sedation, dizziness) in ocinaplon-treated patients did not differ from placebo. These findings indicate that ocinaplon represents a unique approach both for the treatment and understanding of anxiety disorders.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/drug therapy , Pyrimidines/therapeutic use , Receptors, GABA-A/metabolism , Adult , Animals , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Anxiety Disorders/metabolism , Behavior, Animal/drug effects , Conditioning, Operant , Diazepam/pharmacology , Double-Blind Method , Flunitrazepam/metabolism , Germany , Humans , Oocytes/metabolism , Patch-Clamp Techniques , Pentylenetetrazole , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Rats , Saimiri , Tritium , Xenopus laevis
6.
Gene ; 278(1-2): 63-79, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11707323

ABSTRACT

The type B gamma-aminobutryic acid receptor (GABA(B)R) is a G protein coupled receptor that mediates slow pre- and post-synaptic inhibition in the nervous system. We find that the human GABA(B)R2 gene spans greater than 350 kb and contains 2.8 kb of coding region in 19 exons. The overall similarity in genomic structure with regard to conservation of intron position and exon size between human or Drosophila GABA(B)R1 and GABA(B)R2 genes suggests a common ancestral origin. Multiple transcripts GABA(B)R1a-c and GABA(B)R2a-c have been described and alternative splicing has been proposed to result in GABA(B)R1c, GABA(B)R2b and GABA(B)R2c. The results described here provide support for the existence of GABA(B)R1c but not for GABA(B)R2b and GABA(B)R2c. Splice junctions present in the GABA(B)R1 gene sequence are consistent with the formation of GABA(B)R1c by exon skipping of one sushi domain module. The GABA(B)R2 gene lacks canonical splice junctions for the reported variants. Consistent with this, RNA analysis demonstrates the presence of GABA(B)R1c and GABA(B)R2 transcripts in fetal and adult human brain RNA but GABA(B)R2b and GABA(B)R2c transcripts are not detected. These results provide insight into the evolution and transcript diversity of the mammalian GABA(B)R genes.


Subject(s)
Genes/genetics , Receptors, GABA-B/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Brain/embryology , Brain/metabolism , DNA, Complementary/genetics , Drosophila/genetics , Exons , Gene Expression , Gene Expression Regulation, Developmental , Humans , Introns , Molecular Sequence Data , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, GABA/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
7.
Proc Natl Acad Sci U S A ; 97(15): 8600-5, 2000 Jul 18.
Article in English | MEDLINE | ID: mdl-10900018

ABSTRACT

The regulated expression of type A gamma-aminobutyric acid receptor (GABA(A)R) subunit genes is postulated to play a role in neuronal maturation, synaptogenesis, and predisposition to neurological disease. Increases in GABA levels and changes in GABA(A)R subunit gene expression, including decreased beta1 mRNA levels, have been observed in animal models of epilepsy. Persistent exposure to GABA down-regulates GABA(A)R number in primary cultures of neocortical neurons, but the regulatory mechanisms remain unknown. Here, we report the identification of a TATA-less minimal promoter of 296 bp for the human GABA(A)R beta1 subunit gene that is neuron specific and autologously down-regulated by GABA. beta1 promoter activity, mRNA levels, and subunit protein are decreased by persistent GABA(A)R activation. The core promoter, 270 bp, contains an initiator element (Inr) at the major transcriptional start site. Three concatenated copies of the 10-bp Inr and its immediate 3' flanking sequence produce full neural specific activity that is down-regulated by GABA in transiently transfected neocortical neurons. Taking these results together with those of DNase I footprinting, electrophoretic mobility shift analysis, and 2-bp mutagenesis, we conclude that GABA-induced down-regulation of beta1 subunit mRNAs involves the differential binding of a sequence-specific basal transcription factor(s) to the Inr. The results support a transcriptional mechanism for the down-regulation of beta1 subunit GABA(A)R gene expression and raises the possibility that altered levels of sequence-specific basal transcription factors may contribute to neurological disorders such as epilepsy.


Subject(s)
Down-Regulation , Promoter Regions, Genetic , Receptors, GABA-A/genetics , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Cells, Cultured , Hippocampus/cytology , Humans , Molecular Sequence Data , Neocortex/cytology , Neurons/metabolism , Nuclear Proteins/metabolism , Rats , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
8.
J Neurochem ; 74(5): 1858-69, 2000 May.
Article in English | MEDLINE | ID: mdl-10800928

ABSTRACT

The ability of nerve cells to regulate the expression of specific neurotransmitter receptors is of central importance to nervous system function, but little is known about the DNA elements that mediate neuron specific gene expression. The type A gamma-aminobutyric acid (GABA(A)) receptor alpha6-subunit gene, which is expressed exclusively in cerebellar granule cells, presents a unique opportunity to study the cis elements involved in restricting gene expression to a distinct neuronal population. In an effort to identify the regulatory elements that govern cerebellar granule cell-specific gene expression, the proximal 5' flanking regions for the human, rat, and mouse alpha6 genes were cloned and sequenced, and a major transcriptional initiation site was identified in the rodent genes. Functional analysis of rat alpha6 gene-reporter constructs in primary neuronal cultures reveals that a 155-bp TATA-less promoter region (-130 to +25 bp) constitutes a minimal promoter that can drive cerebellar granule cell-specific expression. Internal deletion and decoy competition studies demonstrate that the minimal promoter contains a 60-bp region (-130 to -70 bp) that is critical for enhanced promoter activity in cerebellar granule cells. Activity of the compromised promoter containing the deletion cannot be rescued by placing the 60-bp region downstream of the reporter gene, demonstrating that it is not a classical enhancer but rather a positionally dependent regulator. An additional cerebellar-specific activating sequence is located between -324 and -130 bp, and a downstream negative regulatory region (+158 to +294) has been shown to be active in fibroblasts but inactive in cerebellar granule cells. Taken together, the results suggest a possible mechanism for the control of cerebellar granule cell-specific expression of the GABA(A) receptor alpha6 subunit gene.


Subject(s)
Promoter Regions, Genetic/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Animals , Base Sequence/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Humans , Mice , Molecular Sequence Data , Neurons/physiology , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Substrate Specificity , Transcription, Genetic/physiology
9.
Mol Cell Neurosci ; 13(3): 180-91, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10328880

ABSTRACT

We have identified a gene encoding a GABAB receptor, the human GABABR2, located on chromosome 9q22.1, that is distinct from the recently reported rat GABABR1. GABABR2 structurally resembles GABABR1 (35% identity), having seven transmembrane domains and a large extracellular region, but differs in having a longer carboxy-terminal tail. GABABR2 is localized to the cell surface in transfected COS cells, and negatively couples to adenylyl cyclase in response to GABA, baclofen, and 3-aminopropyl(methyl)phosphinic acid in CHO cells lacking GABABR1. Baclofen action is inhibited by the GABABR antagonist, 2-hydroxysaclofen. The human GABABR2 and GABABR1 genes are differentially expressed in the nervous system, with the greatest difference being detected in the striatum in which GABABR1 but not GABABR2 mRNA transcripts are detected. GABABR2 and GABABR1 mRNAs are also coexpressed in various brain regions such as the Purkinje cell layer of the cerebellum. Identification of a functional homomeric GABABR2 coupled to adenylyl cyclase suggests that the complexity of GABAB pharmacological data is at least in part due to the presence of more than one receptor and opens avenues for future research leading to an understanding of metabotropic GABA receptor signal transduction mechanisms.


Subject(s)
Adenylyl Cyclases/metabolism , Receptors, GABA , Amino Acid Sequence , Animals , Brain/metabolism , CHO Cells/metabolism , COS Cells/metabolism , Cell Membrane/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Cricetinae , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Expressed Sequence Tags , Humans , In Situ Hybridization , Isomerism , Molecular Sequence Data , Rats , Receptors, GABA-B , Retina/metabolism , Transcription, Genetic , Transfection
10.
Gene ; 227(2): 213-22, 1999 Feb 18.
Article in English | MEDLINE | ID: mdl-10023064

ABSTRACT

Nowhere is the record of receptor evolution more accessible than in the organization of the 19 vertebrate genes coding for subunits of the major inhibitory neurotransmitter receptor in the central nervous system, the gamma-aminobutyric acid receptor (GABAAR). Co-expression of alpha, beta, and gamma subunit genes is necessary for the formation of a GABAAR that is potentiated by widely used anxiolytics, anticonvulsants, and hypnotics. The identification of alpha, beta, and gamma genes on chromosomes 4, 5, and 15 suggests that co-localization of a gamma gene with an alpha and beta may be important for brain function. We have now directly examined the organization of GABAAR subunit genes on human chromosomes. Estimates of physical distance using in situ hybridization to cells in interphase, and gene localization using hybridization to cells in metaphase demonstrate the existence of beta-alpha-alpha-gamma gene clusters in cytogenetic bands on chromosomes 4(p12) and 5(q34). Sequencing of PAC clones establishes intercluster conservation of a unique head-to-head configuration for alpha and beta genes on chromosomes 4, 5, and 15. Remarkably, phylogenetic tree analysis predicts the existence of a beta-alpha-gamma ancestral gene cluster in which internal duplication of an ancestral alpha was followed by cluster duplication, resulting in the relative chromosomal positions of modern GABAAR subunit genes in the human genome.


Subject(s)
Genome, Human , Multigene Family/genetics , Receptors, GABA-A/genetics , Chromosome Mapping , Evolution, Molecular , Humans , In Situ Hybridization, Fluorescence , Phylogeny , Polymerase Chain Reaction
11.
Brain Res ; 803(1-2): 153-60, 1998 Aug 24.
Article in English | MEDLINE | ID: mdl-9729352

ABSTRACT

Pregnenolone sulfate (PS) is an abundant neurosteroid that can potentiate or inhibit ligand gated ion channel activity and thereby alter neuronal excitability. Whereas PS is known to inhibit kainate and AMPA responses while potentiating NMDA responses, the dependence of modulation on receptor subunit composition remains to be determined. Toward this end, the effect of PS on recombinant kainate (GluR6), AMPA (GluR1 or GluR3), and NMDA (NR1(100)+NR2A) receptors was characterized electrophysiologically with respect to efficacy and potency of modulation. With Xenopus oocytes expressing GluR1, GluR3 or GluR6 receptors, PS reduces the efficacy of kainate without affecting its potency, indicative of a noncompetitive mechanism of action. Conversely, with oocytes expressing NR1(100)+NR2A subunits, PS enhances the efficacy of NMDA without affecting its potency. Whereas the modulatory efficacy, but not the potency, of PS is increased two-fold by co-injection of NR1(100)+NR2A cRNAs as compared with NR1(100) cRNA alone, there is little or no effect of the NR2A subunit on efficacy or potency of pregnanolone (or epipregnanolone) sulfate as an inhibitor of the NMDA response. This suggests that the NR2A subunit controls the efficacy of neurosteroid enhancement, but not inhibition, which is consistent with our previous finding that potentiating and inhibitory steroids act at distinct sites on the NMDA receptor. This represents a first step towards understanding the role of subunit composition in determining neurosteroid modulation of ionotropic glutamate receptor function.


Subject(s)
Neurotransmitter Agents/pharmacology , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Recombinant Proteins/metabolism , Adrenal Cortex Hormones , Animals , Dose-Response Relationship, Drug , Kainic Acid/antagonists & inhibitors , Kainic Acid/pharmacology , Microinjections , Neurotransmitter Agents/physiology , Oocytes/cytology , Oocytes/drug effects , Pregnenolone/pharmacology , RNA, Complementary/administration & dosage , RNA, Complementary/drug effects , RNA, Complementary/pharmacology , Rats , Receptors, AMPA/drug effects , Receptors, AMPA/genetics , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/drug effects , Xenopus , GluK2 Kainate Receptor
12.
Synapse ; 21(3): 189-274, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8578436

ABSTRACT

The gamma-aminobutyric acid type A (GABAA) receptor represents an elementary switching mechanism integral to the functioning of the central nervous system and a locus for the action of many mood- and emotion-altering agents such as benzodiazepines, barbiturates, steroids, and alcohol. Anxiety, sleep disorders, and convulsive disorders have been effectively treated with therapeutic agents that enhance the action of GABA at the GABAA receptor or increase the concentration of GABA in nervous tissue. The GABAA receptor is a multimeric membrane-spanning ligand-gated ion channel that admits chloride upon binding of the neurotransmitter GABA and is modulated by many endogenous and therapeutically important agents. Since GABA is the major inhibitory neurotransmitter in the CNS, modulation of its response has profound implications for brain functioning. The GABAA receptor is virtually the only site of action for the centrally acting benzodiazepines, the most widely prescribed of the anti-anxiety medications. Increasing evidence points to an important role for GABA in epilepsy and various neuropsychiatric disorders. Recent advances in molecular biology and complementary information derived from pharmacology, biochemistry, electrophysiology, anatomy and cell biology, and behavior have led to a phenomenal growth in our understanding of the structure, function, regulation, and evolution of the GABAA receptor. Benzodiazepines, barbiturates, steroids, polyvalent cations, and ethanol act as positive or negative modulators of receptor function. The description of a receptor gene superfamily comprising the subunits of the GABAA, nicotinic acetylcholine, and glycine receptors has led to a new way of thinking about gene expression and receptor assembly in the nervous system. Seventeen genetically distinct subunit subtypes (alpha 1-alpha 6, beta 1-beta 4, gamma 1-gamma 4, delta, p1-p2) and alternatively spliced variants contribute to the molecular architecture of the GABAA receptor. Mysteriously, certain preferred combinations of subunits, most notably the alpha 1 beta 2 gamma 2 arrangement, are widely codistributed, while the expression of other subunits, such as beta 1 or alpha 6, is severely restricted to specific neurons in the hippocampal formation or cerebellar cortex. Nervous tissue has the capacity to exert control over receptor number, allosteric uncoupling, subunit mRNA levels, and posttranslational modifications through cellular signal transduction mechanisms under active investigation. The genomic organization of the GABAA receptor genes suggests that the present abundance of subtypes arose during evolution through the duplication and translocations of a primordial alpha-beta-gamma gene cluster. This review describes these varied aspects of GABAA receptor research with special emphasis on contemporary cellular and molecular discoveries.


Subject(s)
Genome , Ion Channels/physiology , Receptors, GABA-A/physiology , Animals , Antibodies, Monoclonal , Biological Evolution , Genetic Variation , Humans , Molecular Probes , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , gamma-Aminobutyric Acid/physiology
13.
Genomics ; 26(3): 580-6, 1995 Apr 10.
Article in English | MEDLINE | ID: mdl-7607683

ABSTRACT

We demonstrated previously that an alpha 1-beta 2-gamma 2 gene cluster of the gamma-aminobutyric acid (GABAA) receptor is located on human chromosome 5q34-q35 and that an ancestral alpha-beta-gamma gene cluster probably spawned clusters on chromosomes 4, 5, and 15. Here, we report that the alpha 4 gene (GABRA4) maps to human chromosome 4p14-q12, defining a cluster comprising the alpha 2, alpha 4, beta 1, and gamma 1 genes. The existence of an alpha 2-alpha 4-beta 1-gamma 1 cluster on chromosome 4 and an alpha 1-alpha 6-beta 2-gamma 2 cluster on chromosome 5 provides further evidence that the number of ancestral GABAA receptor subunit genes has been expanded by duplication within an ancestral gene cluster. Moreover, if duplication of the alpha gene occurred before duplication of the ancestral gene cluster, then a heretofore undiscovered subtype of alpha subunit should be located on human chromosome 15q11-q13 within an alpha 5-alpha x-beta 3-gamma 3 gene cluster at the locus for Angelman and Prader-Willi syndromes.


Subject(s)
Chromosomes, Human, Pair 4 , Multigene Family , Receptors, GABA-A/genetics , Amino Acid Sequence , Base Sequence , Biological Evolution , Chromosome Mapping , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 5 , DNA , Humans , Molecular Sequence Data , Polymerase Chain Reaction
15.
Genomics ; 23(3): 528-33, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7851879

ABSTRACT

The gamma-aminobutyric acid receptor (GABAAR) is a multisubunit Cl- channel that mediates most fast inhibitory synaptic transmission in the central nervous system. Molecular evolution has given rise to many genetic variants of GABAAR subunits, including alpha 1-6, beta 1-4, gamma 1-4, delta, and rho 1-2, suggesting that an enormous number of combinations of subunits are possible. Here we report that the beta 2 gene is located on chromosome 5q34-q35, defining a cluster comprising alpha 1, beta 2, and gamma 2 genes that together code for the most abundant GABAAR isoform. The fact that intron position is conserved in the beta 1-3 genes, taken together with the observation that chromosomes 4 and 15 also contain distinct alpha-beta-gamma gene clusters, strongly suggests that an ancestral alpha-beta-gamma cluster was duplicated and translocated to at least two different chromosomes. This organization of GABAAR gene clusters may have been preserved as linkage provides a mechanism for facilitating coordinate gene expression.


Subject(s)
Chromosomes, Human, Pair 5 , Hominidae/genetics , Multigene Family , Receptors, GABA-A/genetics , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Chickens , Chromosome Mapping , Conserved Sequence , DNA Primers , Exons , Humans , Hybrid Cells , Introns , Macromolecular Substances , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Rats , Receptors, GABA-A/biosynthesis , Sequence Homology, Amino Acid
16.
Cell Mol Biol Res ; 39(2): 177-82, 1993.
Article in English | MEDLINE | ID: mdl-8220586

ABSTRACT

It is well known that the ligation of two DNA fragments which are the product of digestion from different restriction enzymes will not lead to the regeneration of either of the original blunt end or cohesive end restriction sites. This property of sequence incompatibility for the original restriction enzyme can be exploited in a general cloning procedure for both PCR products and restricted DNAs. Restriction selection is particularly useful when cloning low abundance polymerase chain reaction (PCR) products and when cloning blunt ended DNA into reporter vectors that lack a method for the selection of recombinants.


Subject(s)
Cloning, Molecular/methods , DNA, Recombinant/isolation & purification , Receptors, GABA/genetics , Base Sequence , DNA Ligases/metabolism , DNA Restriction Enzymes/metabolism , DNA, Recombinant/genetics , Genes , Genetic Vectors , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Substrate Specificity
18.
Psychosom Med ; 52(3): 271-85, 1990.
Article in English | MEDLINE | ID: mdl-2367619

ABSTRACT

A 35-year prospective study was undertaken in 126 former college students to determine the predictive value of psychophysiological patterns previously recorded in response to repetitive laboratory stress experiments. Detailed health information has been obtained in 116 (92.1%) of these subjects. The emotion of "severe anxiety" expressed in one or more of the prior tests appeared to be a reliable marker for increased susceptibility not only to coronary heart disease but to overall future illness. This form of pathological anxiety, moreover, was frequently shown to be linked to marked conflict about hostile impulses. Contrariwise, neither anger-in nor anger-out was found to be associated with a higher incidence of subsequent disease. Failure to express emotion was observed in a variety of subjects who as a group exhibited no predisposition to sickness in later life. Psychological Mastery was predictive of favorable prognosis, but Physiological Mastery, contrary to expectations, did not show statistically significant advantages in that regard. Thus, the construct of "Mastery" itself as a determinant of prognosis was not fully supported by the findings in the present study. Cardiovascular hyperreactivity could not be confirmed as a major biologic mechanism responsible for cardiovascular disease. Such hyperresponses were common in association with "anger-in" without evidence of increased susceptibility to cardiovascular disease or other forms of illness. Further research is needed to identify pathophysiological pathways that may be activated by the emotion of severe anxiety in mediating its apparent relationship with total morbidity and mortality over time.


Subject(s)
Adaptation, Psychological , Arousal , Coronary Disease/psychology , Personality Tests , Psychophysiologic Disorders/psychology , Adult , Alcoholism/psychology , Anger , Anxiety/psychology , Duodenal Ulcer/psychology , Follow-Up Studies , Humans , Hypertension/psychology , Interview, Psychological , Male , Prospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL