Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 14: 1271203, 2023.
Article in English | MEDLINE | ID: mdl-38155909

ABSTRACT

Dementia and autoimmune diseases are prevalent conditions with limited treatment options. Taurine and homotaurine (HT) are naturally occurring sulfonate amino acids, with taurine being highly abundant in animal tissues, but declining with age in the blood. HT is a blood-brain barrier permeable drug under investigation for Alzheimer's disease. HT also has beneficial effects in a mouse model of multiple sclerosis likely through an anti-inflammatory mechanism mediated by GABAA receptor (GABAAR) agonism in immune cells. While both taurine and HT are structural GABA analogs and thought to be GABA mimetics at GABAARs, there is uncertainty concerning their potency as GABA mimetics on native GABAARs. We show that HT is a very potent GABA mimetic, as it evokes GABAAR-mediated currents with an EC50 of 0.4 µM (vs. 3.7 µM for GABA and 116 µM for taurine) in murine cerebellar granule cells in brain slices, with both taurine and HT having similar efficacy in activating native GABAARs. Furthermore, HT displaces the high affinity GABAAR ligand [3H]muscimol at similarly low concentrations (HT IC50 of 0.16 µM vs. 125 µM for taurine) in mouse brain homogenates. The potency of taurine and HT as GABAAR agonists aligns with endogenous concentrations of taurine in the blood and with HT concentrations achieved in the brain following oral administration of HT or the HT pro-drug ALZ-801. Consequently, we discuss that GABAARs subtypes, similar to the ones we studied here in neurons, are plausible targets for mediating the potential beneficial effects of taurine in health and life-span extension and the beneficial HT effects in dementia and autoimmune conditions.

2.
J Neurochem ; 165(3): 445-454, 2023 05.
Article in English | MEDLINE | ID: mdl-36726215

ABSTRACT

Impairment of excretion and enzymatic processing of nitrogen, for example, because of liver or kidney failure, or with urea cycle and creatine synthesis enzyme defects, surprisingly leads to primarily neurologic symptoms, yet the exact mechanisms remain largely mysterious. In guanidinoacetate N-methyltransferase (GAMT) deficiency, the guanidino compound guanidinoacetate (GAA) increases dramatically, including in the cerebrospinal fluid (CSF), and has been implicated in mediating the neurological symptoms in GAMT-deficient patients. GAA is synthesized by arginine-glycine amidinotransferase (AGAT), a promiscuous enzyme that not only transfers the amidino group from arginine to glycine, but also to primary amines in, for example, GABA and taurine to generate γ-guanidinobutyric acid (γ-GBA) and guanidinoethanesulfonic acid (GES), respectively. We show that GAA, γ-GBA, and GES share structural similarities with GABA, evoke GABAA receptor (GABAA R) mediated currents (whereas creatine [methylated GAA] and arginine failed to evoke discernible currents) in cerebellar granule cells in mouse brain slices and displace the high-affinity GABA-site radioligand [3 H]muscimol in total brain homogenate GABAA Rs. While γ-GBA and GES are GABA agonists and displace [3 H]muscimol (EC50 /IC50 between 10 and 40 µM), GAA stands out as particularly potent in both activating GABAA Rs (EC50 ~6 µM) and also displacing the GABAA R ligand [3 H]muscimol (IC50 ~3 µM) at pathophysiologically relevant concentrations. These findings stress the role of substantially elevated GAA as a primary neurotoxic agent in GAMT deficiency and we discuss the potential role of GAA in arginase (and creatine transporter) deficiency which show a much more modest increase in GAA concentrations yet share the unique hyperexcitability neuropathology with GAMT deficiency. We conclude that orthosteric activation of GABAA Rs by GAA, and potentially other GABAA R mimetic guanidino compounds (GCs) like γ-GBA and GES, interferes with normal inhibitory GABAergic neurotransmission which could mediate, and contribute to, neurotoxicity.


Subject(s)
Creatine , Receptors, GABA-A , Mice , Animals , Creatine/pharmacology , Muscimol , Glycine/pharmacology , gamma-Aminobutyric Acid , Arginine
3.
Nat Neurosci ; 24(6): 831-842, 2021 06.
Article in English | MEDLINE | ID: mdl-33820999

ABSTRACT

Social interactions and relationships are often rewarding, but the neural mechanisms through which social interaction drives positive experience remain poorly understood. In this study, we developed an automated operant conditioning system to measure social reward in mice and found that adult mice of both sexes display robust reinforcement of social interaction. Through cell-type-specific manipulations, we identified a crucial role for GABAergic neurons in the medial amygdala (MeA) in promoting the positive reinforcement of social interaction. Moreover, MeA GABAergic neurons mediate social reinforcement behavior through their projections to the medial preoptic area (MPOA) and promote dopamine release in the nucleus accumbens. Finally, activation of this MeA-to-MPOA circuit can robustly overcome avoidance behavior. Together, these findings establish the MeA as a key node for regulating social reward in both sexes, providing new insights into the regulation of social reward beyond the classic mesolimbic reward system.


Subject(s)
Amygdala/physiology , Conditioning, Operant/physiology , Hypothalamus/physiology , Nerve Net/physiology , Reward , Social Behavior , Amygdala/chemistry , Animals , Female , Hypothalamus/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Optogenetics/methods , Reinforcement, Psychology
4.
Neurobiol Dis ; 143: 104977, 2020 09.
Article in English | MEDLINE | ID: mdl-32553709

ABSTRACT

Environmental toxicants have the potential to contribute to the pathophysiology of multiple complex diseases, but the underlying mechanisms remain obscure. One such toxicant is the widely used fungicide ziram, a dithiocarbamate known to have neurotoxic effects and to increase the risk of Parkinson's disease. We have used Drosophila melanogaster as an unbiased discovery tool to identify novel molecular pathways by which ziram may disrupt neuronal function. Consistent with previous results in mammalian cells, we find that ziram increases the probability of synaptic vesicle release by dysregulation of the ubiquitin signaling system. In addition, we find that ziram increases neuronal excitability. Using a combination of live imaging and electrophysiology, we find that ziram increases excitability in both aminergic and glutamatergic neurons. This increased excitability is phenocopied and occluded by null mutant animals of the ether a-go-go (eag) potassium channel. A pharmacological inhibitor of the temperature sensitive hERG (human ether-a-go-go related gene) phenocopies the excitability effects of ziram but only at elevated temperatures. seizure (sei), a fly ortholog of hERG, is thus another candidate target of ziram. Taken together, the eag family of potassium channels emerges as a candidate for mediating some of the toxic effects of ziram. We propose that ziram may contribute to the risk of complex human diseases by blockade of human eag and sei orthologs, such as hERG.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Fungicides, Industrial/toxicity , Neurons/drug effects , Synaptic Vesicles/drug effects , Ziram/toxicity , Animals , Drosophila melanogaster , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Neurons/metabolism , Neurotransmitter Agents/metabolism , Synaptic Vesicles/metabolism
5.
Cell Cycle ; 19(2): 153-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31876231

ABSTRACT

Spinocerebellar ataxias (SCA) are a genetically heterogeneous family of cerebellar neurodegenerative diseases characterized by abnormal firing of Purkinje neurons and degeneration. We recently demonstrated the slowed firing rates seen in several SCAs share a common etiology of hyper-activation of the Src family of non-receptor tyrosine kinases (SFKs). However, the lack of clinically available neuroactive SFK inhibitors lead us to investigate alternative mechanisms to modulate SFK activity. Previous studies demonstrate that SFK activity can be enhanced by the removal of inhibitory phospho-marks by receptor-protein-tyrosine phosphatases (RPTPs). In this Extra View we show that MTSS1 inhibits SFK activity through the binding and inhibition of a subset of the RPTP family members, and lowering RPTP activity in cerebellar slices with peptide inhibitors increases the suppressed Purkinje neuron basal firing rates seen in two different SCA models. Together these results identify RPTPs as novel effectors of Purkinje neuron basal firing, extending the MTSS1/SFK regulatory circuit we previously described and expanding the therapeutic targets for SCA patients.


Subject(s)
Action Potentials/physiology , Protein Tyrosine Phosphatases/metabolism , Purkinje Cells/enzymology , Action Potentials/drug effects , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Mice , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Binding/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Purkinje Cells/drug effects , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/physiopathology
6.
J Neurochem ; 149(1): 41-53, 2019 04.
Article in English | MEDLINE | ID: mdl-30565258

ABSTRACT

Muscimol, the major psychoactive ingredient in the mushroom Amanita muscaria, has been regarded as a universal non-selective GABA-site agonist. Deletion of the GABAA receptor (GABAA R) δ subunit in mice (δKO) leads to a drastic reduction in high-affinity muscimol binding in brain sections and to a lower behavioral sensitivity to muscimol than their wild type counterparts. Here, we use forebrain and cerebellar brain homogenates from WT and δKO mice to show that deletion of the δ subunit leads to a > 50% loss of high-affinity 5 nM [3 H]muscimol-binding sites despite the relatively low abundance of δ-containing GABAA Rs (δ-GABAA R) in the brain. By subtracting residual high-affinity binding in δKO mice and measuring the slow association and dissociation rates we show that native δ-GABAA Rs in WT mice exhibit high-affinity [3 H]muscimol-binding sites (KD ~1.6 nM on α4ßδ receptors in the forebrain and ~1 nM on α6ßδ receptors in the cerebellum at 22°C). Co-expression of the δ subunit with α6 and ß2 or ß3 in recombinant (HEK 293) expression leads to the appearance of a slowly dissociating [3 H]muscimol component. In addition, we compared muscimol currents in recombinant α4ß3δ and α4ß3 receptors and show that δ subunit co-expression leads to highly muscimol-sensitive currents with an estimated EC50 of around 1-2 nM and slow deactivation kinetics. These data indicate that δ subunit incorporation leads to a dramatic increase in GABAA R muscimol sensitivity. We conclude that biochemical and behavioral low-dose muscimol selectivity for δ-subunit-containing receptors is a result of low nanomolar-binding affinity on δ-GABAA Rs.


Subject(s)
Brain/metabolism , Muscimol/metabolism , Receptors, GABA-A/metabolism , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding
7.
Proc Natl Acad Sci U S A ; 115(52): E12407-E12416, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30530649

ABSTRACT

The genetically heterogeneous spinocerebellar ataxias (SCAs) are caused by Purkinje neuron dysfunction and degeneration, but their underlying pathological mechanisms remain elusive. The Src family of nonreceptor tyrosine kinases (SFK) are essential for nervous system homeostasis and are increasingly implicated in degenerative disease. Here we reveal that the SFK suppressor Missing-in-metastasis (MTSS1) is an ataxia locus that links multiple SCAs. MTSS1 loss results in increased SFK activity, reduced Purkinje neuron arborization, and low basal firing rates, followed by cell death. Surprisingly, mouse models for SCA1, SCA2, and SCA5 show elevated SFK activity, with SCA1 and SCA2 displaying dramatically reduced MTSS1 protein levels through reduced gene expression and protein translation, respectively. Treatment of each SCA model with a clinically approved Src inhibitor corrects Purkinje neuron basal firing and delays ataxia progression in MTSS1 mutants. Our results identify a common SCA therapeutic target and demonstrate a key role for MTSS1/SFK in Purkinje neuron survival and ataxia progression.


Subject(s)
Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Animals , Ataxia/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Proteins/metabolism , Purkinje Cells/physiology , Spinocerebellar Ataxias/metabolism , Spinocerebellar Degenerations/metabolism , Spinocerebellar Degenerations/physiopathology , src-Family Kinases/metabolism
8.
Elife ; 62017 05 18.
Article in English | MEDLINE | ID: mdl-28518055

ABSTRACT

Metabotropic glutamate receptor 1 (mGluR1) function in Purkinje neurons (PNs) is essential for cerebellar development and for motor learning and altered mGluR1 signaling causes ataxia. Downstream of mGluR1, dysregulation of calcium homeostasis has been hypothesized as a key pathological event in genetic forms of ataxia but the underlying mechanisms remain unclear. We find in a spinocerebellar ataxia type 2 (SCA2) mouse model that calcium homeostasis in PNs is disturbed across a broad range of physiological conditions. At parallel fiber synapses, mGluR1-mediated excitatory postsynaptic currents (EPSCs) and associated calcium transients are increased and prolonged in SCA2 PNs. In SCA2 PNs, enhanced mGluR1 function is prevented by buffering [Ca2+] at normal resting levels while in wildtype PNs mGluR1 EPSCs are enhanced by elevated [Ca2+]. These findings demonstrate a deleterious positive feedback loop involving elevated intracellular calcium and enhanced mGluR1 function, a mechanism likely to contribute to PN dysfunction and loss in SCA2.


Subject(s)
Calcium/metabolism , Feedback, Physiological , Purkinje Cells/physiology , Receptors, Metabotropic Glutamate/metabolism , Spinocerebellar Ataxias/physiopathology , Animals , Disease Models, Animal , Mice
9.
Hum Mol Genet ; 26(16): 3069-3080, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28525545

ABSTRACT

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominant neurodegenerative disease caused by CAG repeat expansion in the ATXN2 gene. The repeat resides in an encoded region of the gene resulting in polyglutamine (polyQ) expansion which has been assumed to result in gain of function, predominantly, for the ATXN2 protein. We evaluated temporal cerebellar expression profiles by RNA sequencing of ATXN2Q127 mice versus wild-type (WT) littermates. ATXN2Q127 mice are characterized by a progressive motor phenotype onset, and have progressive cerebellar molecular and neurophysiological (Purkinje cell firing frequency) phenotypes. Our analysis revealed previously uncharacterized early and progressive abnormal patterning of cerebellar gene expression. Weighted Gene Coexpression Network Analysis revealed four gene modules that were significantly correlated with disease status, composed primarily of genes associated with GTPase signaling, calcium signaling and cell death. Of these genes, few overlapped with differentially expressed cerebellar genes that we identified in Atxn2-/- knockout mice versus WT littermates, suggesting that loss-of-function is not a significant component of disease pathology. We conclude that SCA2 is a disease characterized by gain of function for ATXN2.


Subject(s)
Gene Regulatory Networks , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Animals , Ataxin-2/genetics , Ataxin-2/metabolism , Ataxins/genetics , Base Sequence , Cerebellum/metabolism , Disease Models, Animal , Gain of Function Mutation , Gene Expression , Gene Expression Profiling , Mice , Mutation , Nerve Tissue Proteins/genetics , Purkinje Cells/metabolism , Sequence Analysis, RNA , Trinucleotide Repeats
10.
Nature ; 544(7650): 362-366, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28405024

ABSTRACT

There are no disease-modifying treatments for adult human neurodegenerative diseases. Here we test RNA-targeted therapies in two mouse models of spinocerebellar ataxia type 2 (SCA2), an autosomal dominant polyglutamine disease. Both models recreate the progressive adult-onset dysfunction and degeneration of a neuronal network that are seen in patients, including decreased firing frequency of cerebellar Purkinje cells and a decline in motor function. We developed a potential therapy directed at the ATXN2 gene by screening 152 antisense oligonucleotides (ASOs). The most promising oligonucleotide, ASO7, downregulated ATXN2 mRNA and protein, which resulted in delayed onset of the SCA2 phenotype. After delivery by intracerebroventricular injection to ATXN2-Q127 mice, ASO7 localized to Purkinje cells, reduced cerebellar ATXN2 expression below 75% for more than 10 weeks without microglial activation, and reduced the levels of cerebellar ATXN2. Treatment of symptomatic mice with ASO7 improved motor function compared to saline-treated mice. ASO7 had a similar effect in the BAC-Q72 SCA2 mouse model, and in both mouse models it normalized protein levels of several SCA2-related proteins expressed in Purkinje cells, including Rgs8, Pcp2, Pcp4, Homer3, Cep76 and Fam107b. Notably, the firing frequency of Purkinje cells returned to normal even when treatment was initiated more than 12 weeks after the onset of the motor phenotype in BAC-Q72 mice. These findings support ASOs as a promising approach for treating some human neurodegenerative diseases.


Subject(s)
Oligonucleotides, Antisense/therapeutic use , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/therapy , Action Potentials , Animals , Ataxin-2/deficiency , Ataxin-2/genetics , Ataxin-2/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Movement , Phenotype , Purkinje Cells/metabolism , Purkinje Cells/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rotarod Performance Test , Spinocerebellar Ataxias/pathology , Spinocerebellar Ataxias/physiopathology
11.
J Physiol ; 594(16): 4653-60, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27198167

ABSTRACT

Degenerative ataxias are a common form of neurodegenerative disease that affect about 20 individuals per 100,000. The autosomal dominant spinocerebellar ataxias (SCAs) are caused by a variety of protein coding mutations (single nucleotide changes, deletions and expansions) in single genes. Affected genes encode plasma membrane and intracellular ion channels, membrane receptors, protein kinases, protein phosphatases and proteins of unknown function. Although SCA-linked genes are quite diverse they share two key features: first, they are highly, although not exclusively, expressed in cerebellar Purkinje neurons (PNs), and second, when mutated they lead ultimately to the degeneration of PNs. In this review we summarize ataxia-related changes in PN neurophysiology that have been observed in various mouse knockout lines and in transgenic models of human SCA. We also highlight emerging evidence that altered metabotropic glutamate receptor signalling and disrupted calcium homeostasis in PNs form a common, early pathophysiological mechanism in SCAs. Together these findings indicate that aberrant calcium signalling and profound changes in PN neurophysiology precede PN cell loss and are likely to lead to cerebellar circuit dysfunction that explains behavioural signs of ataxia characteristic of the disease.


Subject(s)
Spinocerebellar Ataxias/physiopathology , Action Potentials , Animals , Calcium/physiology , Humans , Purkinje Cells/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction
12.
Sci Transl Med ; 5(208): 208ra149, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24154603

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative condition characterized by loss of motor neurons in the brain and spinal cord. Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9ORF72 gene are the most common cause of the familial form of ALS (C9-ALS), as well as frontotemporal lobar degeneration and other neurological diseases. How the repeat expansion causes disease remains unclear, with both loss of function (haploinsufficiency) and gain of function (either toxic RNA or protein products) proposed. We report a cellular model of C9-ALS with motor neurons differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying the C9ORF72 repeat expansion. No significant loss of C9ORF72 expression was observed, and knockdown of the transcript was not toxic to cultured human motor neurons. Transcription of the repeat was increased, leading to accumulation of GGGGCC repeat-containing RNA foci selectively in C9-ALS iPSC-derived motor neurons. Repeat-containing RNA foci colocalized with hnRNPA1 and Pur-α, suggesting that they may be able to alter RNA metabolism. C9-ALS motor neurons showed altered expression of genes involved in membrane excitability including DPP6, and demonstrated a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons. Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in C9-ALS motor neurons. These data show that patient-derived motor neurons can be used to delineate pathogenic events in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA Repeat Expansion/genetics , Induced Pluripotent Stem Cells/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Proteins/genetics , RNA/metabolism , C9orf72 Protein , Exons/genetics , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , RNA/biosynthesis , RNA/genetics , Transcription, Genetic/drug effects
13.
PLoS One ; 8(8): e72976, 2013.
Article in English | MEDLINE | ID: mdl-23977374

ABSTRACT

GABAA receptors (GABARs) are the targets of a wide variety of modulatory drugs which enhance chloride flux through GABAR ion channels. Certain GABAR modulators appear to acutely enhance the function of δ subunit-containing GABAR subtypes responsible for tonic forms of inhibition. Here we identify a reinforcing circuit mechanism by which these drugs, in addition to directly enhancing GABAR function, also increase GABA release. Electrophysiological recordings in cerebellar slices from rats homozygous for the ethanol-hypersensitive (α6100Q) allele show that modulators and agonists selective for δ-containing GABARs such as THDOC, ethanol and THIP (gaboxadol) increased the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in granule cells. Ethanol fails to augment granule cell sIPSC frequency in the presence of glutamate receptor antagonists, indicating that circuit mechanisms involving granule cell output contribute to ethanol-enhancement of synaptic inhibition. Additionally, GABAR antagonists decrease ethanol-induced enhancement of Golgi cell firing. Consistent with a role for glutamatergic inputs, THIP-induced increases in Golgi cell firing are abolished by glutamate receptor antagonists. Moreover, THIP enhances the frequency of spontaneous excitatory postsynaptic currents in Golgi cells. Analyses of knockout mice indicate that δ subunit-containing GABARs are required for enhancing GABA release in the presence of ethanol and THIP. The limited expression of the GABAR δ subunit protein within the cerebellar cortex suggests that an indirect, circuit mechanism is responsible for stimulating Golgi cell GABA release by drugs selective for extrasynaptic isoforms of GABARs. Such circuit effects reinforce direct actions of these positive modulators on tonic GABAergic inhibition and are likely to contribute to the potent effect of these compounds as nervous system depressants.


Subject(s)
Cerebellum/cytology , GABA-A Receptor Agonists/pharmacology , Neural Inhibition/drug effects , Neurons/metabolism , Synapses/metabolism , Action Potentials/drug effects , Animals , Ethanol/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glutamates/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Ion Channel Gating/drug effects , Isoxazoles/pharmacology , Mice, Inbred C57BL , Neurons/drug effects , Protein Subunits/metabolism , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, Glutamate/metabolism , Synapses/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
14.
Hum Mol Genet ; 22(2): 271-83, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23087021

ABSTRACT

Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominantly inherited disorder, which is caused by a pathological expansion of a polyglutamine (polyQ) tract in the coding region of the ATXN2 gene. Like other ataxias, SCA2 most overtly affects Purkinje cells (PCs) in the cerebellum. Using a transgenic mouse model expressing a full-length ATXN2(Q127)-complementary DNA under control of the Pcp2 promoter (a PC-specific promoter), we examined the time course of behavioral, morphologic, biochemical and physiological changes with particular attention to PC firing in the cerebellar slice. Although motor performance began to deteriorate at 8 weeks of age, reductions in PC number were not seen until after 12 weeks. Decreases in the PC firing frequency first showed at 6 weeks and paralleled deterioration of motor performance with progression of disease. Transcription changes in several PC-specific genes such as Calb1 and Pcp2 mirrored the time course of changes in PC physiology with calbindin-28 K changes showing the first small, but significant decreases at 4 weeks. These results emphasize that in this model of SCA2, physiological and behavioral phenotypes precede morphological changes by several weeks and provide a rationale for future studies examining the effects of restoration of firing frequency on motor function and prevention of future loss of PCs.


Subject(s)
Gene Expression , Psychomotor Performance , Purkinje Cells/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Animals , Ataxins , Cerebellum/metabolism , Cerebellum/pathology , Cerebellum/physiopathology , Disease Models, Animal , Disease Progression , Female , Male , Mice , Mice, Transgenic , Motor Activity/genetics , Mutation , Nerve Tissue Proteins/genetics , Phenotype , Purkinje Cells/pathology , RNA Splicing , RNA-Binding Proteins/metabolism , Time Factors
15.
Genes Dev ; 26(5): 445-60, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22357600

ABSTRACT

The Rbfox proteins (Rbfox1, Rbfox2, and Rbfox3) regulate the alternative splicing of many important neuronal transcripts and have been implicated in a variety of neurological disorders. However, their roles in brain development and function are not well understood, in part due to redundancy in their activities. Here we show that, unlike Rbfox1 deletion, the CNS-specific deletion of Rbfox2 disrupts cerebellar development. Genome-wide analysis of Rbfox2(-/-) brain RNA identifies numerous splicing changes altering proteins important both for brain development and mature neuronal function. To separate developmental defects from alterations in the physiology of mature cells, Rbfox1 and Rbfox2 were deleted from mature Purkinje cells, resulting in highly irregular firing. Notably, the Scn8a mRNA encoding the Na(v)1.6 sodium channel, a key mediator of Purkinje cell pacemaking, is improperly spliced in RbFox2 and Rbfox1 mutant brains, leading to highly reduced protein expression. Thus, Rbfox2 protein controls a post-transcriptional program required for proper brain development. Rbfox2 is subsequently required with Rbfox1 to maintain mature neuronal physiology, specifically Purkinje cell pacemaking, through their shared control of sodium channel transcript splicing.


Subject(s)
Cerebellum/embryology , Motor Neurons/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Cerebellum/cytology , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Mice , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism , RNA Splicing/genetics , RNA Splicing Factors , Sodium Channels/metabolism
16.
J Neurophysiol ; 106(4): 2057-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795619

ABSTRACT

Extrasynaptic GABA(A) receptors (eGABARs) allow ambient GABA to tonically regulate neuronal excitability and are implicated as targets for ethanol and anesthetics. These receptors are thought to be heteropentameric proteins made up of two α subunits-either α4 or α6-two ß2 or ß3 subunits, and one δ subunit. The GABA analog 4,5,6,7-tetrahydroisoxazolo (5,4-c)pyridin-3(-ol) (THIP) has been proposed as a selective ligand for eGABARs. Behavioral and in vitro studies suggest that eGABARs have nanomolar affinity for THIP; however, all published studies on recombinant versions of eGABARs report micromolar affinities. Here, we examine THIP sensitivity of native eGABARs on cerebellar neurons and on reconstituted GABARs in heterologous systems. Concentration-response data for THIP, obtained from cerebellar granule cells and molecular layer interneurons in wild-type and δ subunit knockout slices, confirm that submicromolar THIP sensitivity requires δ subunits. In recombinant experiments, we find that δ subunit coexpression leads to receptors activated by nanomolar THIP concentrations (EC(50) of 30-50 nM for α4ß3δ and α6ß3δ), a sensitivity almost 1,000-fold higher than receptors formed by α4/6 and ß3 subunits. In contrast, γ2 subunit expression significantly reduces THIP sensitivity. Even when δ subunit cDNA or cRNA was supplied in excess, high- and low-sensitivity THIP responses were often apparent, indicative of variable mixtures of low-affinity αß and high-affinity αßδ receptors. We conclude that δ subunit incorporation into GABARs leads to a dramatic increase in THIP sensitivity, a defining feature that accounts for the unique behavioral and neurophysiological properties of THIP.


Subject(s)
GABA Agonists/pharmacology , GABAergic Neurons/drug effects , Isoxazoles/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Animals , Cerebellar Cortex/cytology , Dose-Response Relationship, Drug , GABA Agonists/administration & dosage , GABA Agonists/pharmacokinetics , GABAergic Neurons/physiology , HEK293 Cells , Humans , Isoxazoles/administration & dosage , Isoxazoles/pharmacokinetics , Mice , Mice, Knockout , Oocytes , Patch-Clamp Techniques , Protein Isoforms/physiology , Protein Multimerization , Protein Subunits , Receptors, GABA-A/chemistry , Receptors, GABA-A/deficiency , Receptors, GABA-A/genetics , Recombinant Fusion Proteins/physiology , Solubility , Xenopus laevis
17.
Mol Pharmacol ; 78(5): 918-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20699325

ABSTRACT

GABA(A) receptors (GABA(A)Rs) have long been a focus as targets for alcohol actions. Recent work suggests that tonic GABAergic inhibition mediated by extrasynaptic δ subunit-containing GABA(A)Rs is uniquely sensitive to ethanol and enhanced at concentrations relevant for human alcohol consumption. Ethanol enhancement of recombinant α4ß3δ receptors is blocked by the behavioral alcohol antagonist 8-azido-5,6-dihydro-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylic acid ethyl ester (Ro15-4513), suggesting that EtOH/Ro15-4513-sensitive receptors mediate important behavioral alcohol actions. Here we confirm alcohol/alcohol antagonist sensitivity of α4ß3δ receptors using human clones expressed in a human cell line and test the hypothesis that discrepant findings concerning the high alcohol sensitivity of these receptors are due to difficulties incorporating δ subunits into functional receptors. To track δ subunit incorporation, we used a functional tag, a single amino acid change (H68A) in a benzodiazepine binding residue in which a histidine in the δ subunit is replaced by an alanine residue found at the homologous position in γ subunits. We demonstrate that the δH68A substitution confers diazepam sensitivity to otherwise diazepam-insensitive α4ß3δ receptors. The extent of enhancement of α4ß3δH68A receptors by 1 µM diazepam, 30 mM EtOH, and 1 µM ß-carboline-3-carboxy ethyl ester (but not 1 µM Zn(2+) block) is correlated in individual recordings, suggesting that δ subunit incorporation into recombinant GABA(A)Rs varies from cell to cell and that this variation accounts for the variable pharmacological profile. These data are consistent with the notion that δ subunit-incorporation is often incomplete in recombinant systems yet is necessary for high ethanol sensitivity, one of the features of native δ subunit-containing GABA(A)Rs.


Subject(s)
Azides/pharmacology , Benzodiazepines/pharmacology , Ethanol/antagonists & inhibitors , Ethanol/pharmacology , Receptors, GABA-A/biosynthesis , Allosteric Regulation , Animals , Carbolines/pharmacology , Cations, Divalent , Cell Line , Diazepam/pharmacology , Humans , Mutation , Patch-Clamp Techniques , Protein Subunits/biosynthesis , Protein Subunits/genetics , Rats , Receptors, GABA-A/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection , Zinc/pharmacology
18.
Neuropharmacology ; 56(1): 155-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18778723

ABSTRACT

General anesthetics, once thought to exert their effects through non-specific membrane effects, have highly specific ion channel targets that can silence neuronal populations in the nervous system, thereby causing unconsciousness and immobility, characteristic of general anesthesia. Inhibitory GABA(A) receptors (GABA(A)Rs), particularly highly GABA-sensitive extrasynaptic receptor subtypes that give rise to sustained inhibitory currents, are uniquely sensitive to GABA(A)R-active anesthetics. A prominent population of extrasynaptic GABA(A)Rs is made up of alpha4, beta2 or beta3, and delta subunits. Considering the demonstrated importance of GABA receptor beta3 subunits for in vivo anesthetic effects of etomidate and propofol, we decided to investigate the effects of GABA anesthetics on "extrasynaptic" alpha4beta3delta and also binary alpha4beta3 receptors expressed in human embryonic kidney (HEK) cells. Consistent with previous work on similar receptor subtypes we show that maximal GABA currents through "extrasynaptic" alpha4beta3delta receptors, receptors defined by sensitivity to EtOH (30mM) and the beta-carboline beta-CCE (1microM), are enhanced by the GABA(A)R-active anesthetics etomidate, propofol, and the neurosteroid anesthetic THDOC. Furthermore, we show that receptors formed by alpha4beta3 subunits alone also show high GABA sensitivity and that saturating GABA responses of alpha4beta3 receptors are increased to the same extent by etomidate, propofol, and THDOC as are alpha4beta3delta receptors. Therefore, both alpha4beta3 and alpha4beta3delta receptors show low GABA efficacy, and GABA is also a partial agonist on certain binary alphabeta receptor subtypes. Increasing GABA efficacy on alpha4/6beta3delta and alpha4beta3 receptors is likely to make an important contribution to the anesthetic effects of etomidate, propofol and the neurosteroid THDOC.


Subject(s)
Gene Expression/drug effects , Hypnotics and Sedatives/pharmacology , Receptors, GABA-A/biosynthesis , Recombinant Proteins/biosynthesis , gamma-Aminobutyric Acid/pharmacology , Cell Line, Transformed , Central Nervous System Depressants/pharmacology , Desoxycorticosterone/analogs & derivatives , Desoxycorticosterone/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Electric Stimulation/methods , Ethanol/pharmacology , Etomidate/pharmacology , Humans , Patch-Clamp Techniques/methods , Propofol/pharmacology , Transfection/methods
19.
Alcohol ; 41(3): 201-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17591543

ABSTRACT

This review discusses evidence for and apparent controversy about, gamma-aminobutyric acid type A (GABAA) receptor (GABAAR) subtypes that mediate alcohol effects experienced during social drinking. GABAARs that contain the beta3 and delta subunits were shown to be enhanced by alcohol concentrations that mirror the concentration dependence of alcohol responses in humans. A mutation (alpha6R100Q) previously found in alcohol nontolerant rats in the cerebellar GABAAR alpha6 subunit is sufficient for increased alcohol-induced ataxia in rats homozygous for this mutation (alpha6-100QQ) and further increases alcohol sensitivity of tonic GABA currents (mediated by alpha6betadelta receptors) in cerebellar granule cells of alpha6-100QQ rats and in recombinant alpha6R100Qbeta3delta receptors. This provided the first direct evidence that these types of receptors mediate behavioral effects of ethanol. Furthermore, the behavioral alcohol antagonist Ro15-4513 specifically reverses ethanol enhancement on alpha4/6beta3delta receptors. Unexpectedly, native and recombinant alpha4/6beta3delta receptors bind the behavioral alcohol antagonist Ro15-4513 with high affinity and this binding is competitive with EtOH, suggesting a specific and mutually exclusive (competitive) ethanol/Ro15-4513 site, which explains the puzzling activity of Ro15-4513 as a behavioral alcohol antagonist. Our conclusion from these findings is that alcohol/Ro15-4513-sensitive GABAAR subtypes are important alcohol targets and that alcohol at relevant concentrations is more specific than previously thought. In this review, we discuss technical difficulties in expressing recombinant delta subunit-containing receptors in oocytes and mammalian cells that may have contributed to negative results and confusion. Not only because we have reproduced detailed positive results numerous times, and we and many others have built extensively on basic findings, but also because we explain and combine many previously puzzling results into a coherent and highly plausible paradigm on how alcohol exerts an important part of its action in the brain, we are confident about our findings and conclusions. However, many important open questions remain to be answered.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, GABA-A/drug effects , Wine , Animals , Azides/pharmacology , Benzodiazepines/pharmacology , Humans
20.
Proc Natl Acad Sci U S A ; 103(22): 8546-51, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16581914

ABSTRACT

Although GABA(A) receptors have long been implicated in mediating ethanol (EtOH) actions, receptors containing the "nonsynaptic" delta subunit only recently have been shown to be uniquely sensitive to EtOH. Here, we show that delta subunit-containing receptors bind the imidazo-benzodiazepines (BZs) flumazenil and Ro15-4513 with high affinity (K(d) < 10 nM), contrary to the widely held belief that these receptors are insensitive to BZs. In immunopurified native cerebellar and recombinant delta subunit-containing receptors, binding of the alcohol antagonist [(3)H]Ro15-4513 is inhibited by low concentrations of EtOH (K(i) approximately 8 mM). Also, Ro15-4513 binding is inhibited by BZ-site ligands that have been shown to reverse the behavioral alcohol antagonism of Ro15-4513 (i.e., flumazenil, beta-carbolinecarboxylate ethyl ester (beta-CCE), and N-methyl-beta-carboline-3-carboxamide (FG7142), but not including any classical BZ agonists like diazepam). Experiments that were designed to distinguish between a competitive and allosteric mechanism suggest that EtOH and Ro15-4513 occupy a mutually exclusive binding site. The fact that only Ro15-4513, but not flumazenil, can inhibit the EtOH effect, and that Ro15-4513 differs from flumazenil by only a single group in the molecule (an azido group at the C7 position of the BZ ring) suggest that this azido group in Ro15-4513 might be the area that overlaps with the alcohol-binding site. Our findings, combined with previous observations that Ro15-4513 is a behavioral alcohol antagonist, suggest that many of the behavioral effects of EtOH at relevant physiological concentrations are mediated by EtOH/Ro15-4513-sensitive GABA(A) receptors.


Subject(s)
Azides/antagonists & inhibitors , Azides/metabolism , Benzodiazepines/antagonists & inhibitors , Benzodiazepines/metabolism , Ethanol/pharmacology , GABA-A Receptor Antagonists , Receptors, GABA-A/metabolism , Animals , Azides/chemistry , Azides/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Binding, Competitive/drug effects , Brain/drug effects , Brain/metabolism , Cell Line , Humans , Ligands , Molecular Structure , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, GABA-A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...