Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 29(23): 3744-3756, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33084871

ABSTRACT

Several X-linked neurodevelopmental disorders including Rett syndrome, induced by mutations in the MECP2 gene, and fragile X syndrome (FXS), caused by mutations in the FMR1 gene, share autism-related features. The mRNA coding for methyl CpG binding protein 2 (MeCP2) has previously been identified as a substrate for the mRNA-binding protein, fragile X mental retardation protein (FMRP), which is silenced in FXS. Here, we report a homeostatic relationship between these two key regulators of gene expression in mouse models of FXS (Fmr1 Knockout (KO)) and Rett syndrome (MeCP2 KO). We found that the level of MeCP2 protein in the cerebral cortex was elevated in Fmr1 KO mice, whereas MeCP2 KO mice displayed reduced levels of FMRP, implicating interplay between the activities of MeCP2 and FMRP. Indeed, knockdown of MeCP2 with short hairpin RNAs led to a reduction of FMRP in mouse Neuro2A and in human HEK-293 cells, suggesting a reciprocal coupling in the expression level of these two regulatory proteins. Intra-cerebroventricular injection of an adeno-associated viral vector coding for FMRP led to a concomitant reduction in MeCP2 expression in vivo and partially corrected locomotor hyperactivity. Additionally, the level of MeCP2 in the posterior cortex correlated with the severity of the hyperactive phenotype in Fmr1 KO mice. These results demonstrate that MeCP2 and FMRP operate within a previously undefined homeostatic relationship. Our findings also suggest that MeCP2 overexpression in Fmr1 KO mouse posterior cerebral cortex may contribute to the fragile X locomotor hyperactivity phenotype.


Subject(s)
Cerebral Cortex/pathology , Disease Models, Animal , Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/pathology , Gene Expression Regulation , Methyl-CpG-Binding Protein 2/physiology , Phenotype , Animals , Cerebral Cortex/metabolism , Female , Fragile X Syndrome/etiology , Fragile X Syndrome/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Mol Psychiatry ; 25(9): 2017-2035, 2020 09.
Article in English | MEDLINE | ID: mdl-30224722

ABSTRACT

Principal neurons encode information by varying their firing rate and patterns precisely fine-tuned through GABAergic interneurons. Dysregulation of inhibition can lead to neuropsychiatric disorders, yet little is known about the molecular basis underlying inhibitory control. Here, we find that excessive GABA release from basket cells (BCs) attenuates the firing frequency of Purkinje neurons (PNs) in the cerebellum of Fragile X Mental Retardation 1 (Fmr1) knockout (KO) mice, a model of Fragile X Syndrome (FXS) with abrogated expression of the Fragile X Mental Retardation Protein (FMRP). This over-inhibition originates from increased excitability and Ca2+ transients in the presynaptic terminals, where Kv1.2 potassium channels are downregulated. By paired patch-clamp recordings, we further demonstrate that acutely introducing an N-terminal fragment of FMRP into BCs normalizes GABA release in the Fmr1-KO synapses. Conversely, direct injection of an inhibitory FMRP antibody into BCs, or membrane depolarization of BCs, enhances GABA release in the wild type synapses, leading to abnormal inhibitory transmission comparable to the Fmr1-KO neurons. We discover that the N-terminus of FMRP directly binds to a phosphorylated serine motif on the C-terminus of Kv1.2; and that loss of this interaction in BCs exaggerates GABA release, compromising the firing activity of PNs and thus the output from the cerebellar circuitry. An allosteric Kv1.2 agonist, docosahexaenoic acid, rectifies the dysregulated inhibition in vitro as well as acoustic startle reflex and social interaction in vivo of the Fmr1-KO mice. Our results unravel a novel molecular locus for targeted intervention of FXS and perhaps autism.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Interneurons/metabolism , Mice , Mice, Knockout , Synaptic Transmission , gamma-Aminobutyric Acid
3.
Neurobiol Dis ; 146: 105118, 2020 12.
Article in English | MEDLINE | ID: mdl-33031903

ABSTRACT

Fragile X syndrome (FXS), a neurodevelopmental disorder with autistic features, is caused by the loss of the fragile X mental retardation protein. Sex-specific differences in the clinical profile have been observed in FXS patients, but few studies have directly compared males and females in rodent models of FXS. To address this, we performed electroencephalography (EEG) recordings and a battery of autism-related behavioral tasks on juvenile and young adult Fmr1 knockout (KO) rats. EEG analysis demonstrated that compared to wild-type, male Fmr1 KO rats showed an increase in gamma frequency band power in the frontal cortex during the sleep-like immobile state, and both male and female KO rats failed to show an increase in delta frequency power in the sleep-like state, as observed in wild-type rats. Previous studies of EEG profiles in FXS subjects also reported abnormally increased gamma frequency band power, highlighting this parameter as a potential translatable biomarker. Both male and female Fmr1 KO rats displayed reduced exploratory behaviors in the center zone of the open field test, and increased distance travelled in an analysis of 24-h home cage activity, an effect that was more prominent during the nocturnal phase. Reduced wins against wild-type opponents in the tube test of social dominance was seen in both sexes. In contrast, increased repetitive behaviors in the wood chew test was observed in male but not female KO rats, while increased freezing in a fear conditioning test was observed only in the female KO rats. Our findings highlight sex differences between male and female Fmr1 KO rats, and indicate that the rat model of FXS could be a useful tool for the development of new therapeutics for treating this debilitating neurodevelopmental disorder.


Subject(s)
Auditory Cortex/physiopathology , Autistic Disorder/physiopathology , Behavior, Animal/physiology , Fragile X Syndrome/physiopathology , Acoustic Stimulation/methods , Animals , Anxiety/physiopathology , Auditory Cortex/metabolism , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Disease Models, Animal , Electroencephalography/methods , Exploratory Behavior/physiology , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Rats
4.
Am J Pathol ; 187(4): 864-883, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28342444

ABSTRACT

Farber disease is a rare autosomal recessive disorder caused by acid ceramidase deficiency that usually presents as early-onset progressive visceral and neurologic disease. To understand the neurologic abnormality, we investigated behavioral, biochemical, and cellular abnormalities in the central nervous system of Asah1P361R/P361R mice, which serve as a model of Farber disease. Behaviorally, the mutant mice had reduced voluntary locomotion and exploration, increased thigmotaxis, abnormal spectra of basic behavioral activities, impaired muscle grip strength, and defects in motor coordination. A few mutant mice developed hydrocephalus. Mass spectrometry revealed elevations of ceramides, hydroxy-ceramides, dihydroceramides, sphingosine, dihexosylceramides, and monosialodihexosylganglioside in the brain. The highest accumulation was in hydroxy-ceramides. Storage compound distribution was analyzed by mass spectrometry imaging and morphologic analyses and revealed involvement of a wide range of central nervous system cell types (eg, neurons, endothelial cells, and choroid plexus cells), most notably microglia and/or macrophages. Coalescing and mostly perivascular granuloma-like accumulations of storage-laden CD68+ microglia and/or macrophages were seen as early as 3 weeks of age and located preferentially in white matter, periventricular zones, and meninges. Neurodegeneration was also evident in specific cerebral areas in late disease. Overall, our central nervous system studies in Asah1P361R/P361R mice substantially extend the understanding of human Farber disease and suggest that this model can be used to advance therapeutic approaches for this currently untreatable disorder.


Subject(s)
Central Nervous System/abnormalities , Farber Lipogranulomatosis/complications , Farber Lipogranulomatosis/pathology , Nervous System Malformations/etiology , Nervous System Malformations/pathology , Acid Ceramidase/metabolism , Animals , Behavior, Animal , Central Nervous System/pathology , Cerebellum/pathology , Cerebellum/ultrastructure , Cerebrum/pathology , Cerebrum/ultrastructure , Homozygote , Hydrocephalus/pathology , Mice , Mice, Transgenic , Motor Activity , Neurons/pathology , Neurons/ultrastructure , Phenotype , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sphingolipids/metabolism , Time Factors
5.
Hum Mol Genet ; 22(19): 3920-30, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23740941

ABSTRACT

Fragile X Syndrome is the most common inherited cause of autism. Fragile X mental retardation protein (FMRP), which is absent in fragile X, is an mRNA binding protein that regulates the translation of hundreds of different mRNA transcripts. In the adult brain, FMRP is expressed primarily in the neurons; however, it is also expressed in developing glial cells, where its function is not well understood. Here, we show that fragile X (Fmr1) knockout mice display abnormalities in the myelination of cerebellar axons as early as the first postnatal week, corresponding roughly to the equivalent time in human brain development when symptoms of the syndrome first become apparent (1-3 years of age). At postnatal day (PND) 7, diffusion tensor magnetic resonance imaging showed reduced volume of the Fmr1 cerebellum compared with wild-type mice, concomitant with an 80-85% reduction in the expression of myelin basic protein, fewer myelinated axons and reduced thickness of myelin sheaths, as measured by electron microscopy. Both the expression of the proteoglycan NG2 and the number of PDGFRα+/NG2+ oligodendrocyte precursor cells were reduced in the Fmr1 cerebellum at PND 7. Although myelin proteins were still depressed at PND 15, they regained wild-type levels by PND 30. These findings suggest that impaired maturation or function of oligodendrocyte precursor cells induces delayed myelination in the Fmr1 mouse brain. Our results bolster an emerging recognition that white matter abnormalities in early postnatal brain development represent an underlying neurological deficit in Fragile X syndrome.


Subject(s)
2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Cerebellum/physiopathology , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/physiopathology , Myelin Sheath/physiology , 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics , Animals , Animals, Newborn , Cerebellum/growth & development , Cerebellum/metabolism , Cerebellum/pathology , Disease Models, Animal , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/pathology , Neurons/physiology , Oligodendroglia/cytology
6.
Mol Ther Methods Clin Dev ; 28: 330-343, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36874244

ABSTRACT

Impairment of GABAergic inhibitory neuronal function is linked to epilepsy and other neurological and psychiatric disorders. Recombinant adeno-associated virus (rAAV)-based gene therapy targeting GABAergic neurons is a promising treatment for GABA-associated disorders. However, there is a need to develop rAAV-compatible gene-regulatory elements capable of selectively driving expression in GABAergic neurons throughout the brain. Here, we designed several novel GABAergic gene promoters. In silico analyses, including evolutionarily conserved DNA sequence alignments and transcription factor binding site searches among GABAergic neuronal genes, were carried out to reveal novel sequences for use as rAAV-compatible promoters. rAAVs (serotype 9) were injected into the CSF of neonatal mice and into the brain parenchyma of adult mice to assess promoter specificity. In mice injected neonatally, transgene expression was detected in multiple brain regions with very high neuronal specificity and moderate-to-high GABAergic neuronal selectivity. The GABA promoters differed greatly in their levels of expression and, in some brain regions, showed strikingly different patterns of GABAergic neuron transduction. This study is the first report of rAAV vectors that are functional in multiple brain regions using promoters designed by in silico analyses from multiple GABAergic genes. These novel GABA-targeting vectors may be useful tools to advance gene therapy for GABA-associated disorders.

7.
JCI Insight ; 8(11)2023 06 08.
Article in English | MEDLINE | ID: mdl-37288657

ABSTRACT

Fragile X syndrome is a neurodevelopmental disorder caused by the absence of the mRNA-binding protein fragile X messenger ribonucleoprotein (FMRP). Because FMRP is a highly pleiotropic protein controlling the expression of hundreds of genes, viral vector-mediated gene replacement therapy is viewed as a potential viable treatment to correct the fundamental underlying molecular pathology inherent in the disorder. Here, we studied the safety profile and therapeutic effects of a clinically relevant dose of a self-complementary adeno-associated viral (AAV) vector containing a major human brain isoform of FMRP after intrathecal injection into wild-type and fragile X-KO mice. Analysis of the cellular transduction in the brain indicated primarily neuronal transduction with relatively sparse glial expression, similar to endogenous FMRP expression in untreated wild-type mice. AAV vector-treated KO mice showed recovery from epileptic seizures, normalization of fear conditioning, reversal of slow-wave deficits as measured via electroencephalographic recordings, and restoration of abnormal circadian motor activity and sleep. Further assessment of vector efficacy by tracking and analyzing individual responses demonstrated correlations between the level and distribution of brain transduction and drug response. These preclinical findings further demonstrate the validity of AAV vector-mediated gene therapy for treating the most common genetic cause of cognitive impairment and autism in children.


Subject(s)
Fear , Fragile X Mental Retardation Protein , Animals , Humans , Mice , Fragile X Mental Retardation Protein/genetics , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Seizures/genetics , Seizures/therapy
8.
J Biol Chem ; 286(47): 40922-33, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21969374

ABSTRACT

The calcium-sensing receptor (CaSR) is a family C G protein-coupled receptor that is activated by elevated levels of extracellular divalent cations. The CaSR couples to members of the G(q) family of G proteins, and in the endocrine system this receptor is instrumental in regulating the release of parathyroid hormone from the parathyroid gland and calcitonin from thyroid cells. Here, we demonstrate that in medullary thyroid carcinoma cells, the CaSR promotes cellular adhesion and migration via coupling to members of the integrin family of extracellular matrix-binding proteins. Immunopurification and mass spectrometry, co-immunoprecipitation, and co-localization studies showed that the CaSR and ß1-containing integrins are components of a macromolecular protein complex. In fibronectin-based cell adhesion and migration assays, the CaSR-positive allosteric modulator NPS R-568 induced a concentration-dependent increase in cell adhesion and migration; both of these effects were blocked by a specific CaSR-negative allosteric modulator. These effects were mediated by integrins because they were blocked by a peptide inhibitor of integrin binding to fibronectin and ß1 knockdown experiments. An analysis of intracellular signaling pathways revealed a key role for CaSR-induced phospholipase C activation and the release of intracellular calcium. These results demonstrate for the first time that an ion-sensing G protein-coupled receptor functionally couples to the integrins and, in conjunction with intracellular calcium release, promotes cellular adhesion and migration in tumor cells. The significance of this interaction is further highlighted by studies implicating the CaSR in cancer metastasis, axonal growth, and stem cell attachment, functions that rely on integrin-mediated cell adhesion.


Subject(s)
Cell Movement , Integrins/metabolism , Receptors, Calcium-Sensing/metabolism , Allosteric Regulation/drug effects , Aniline Compounds/pharmacology , Animals , Calcium/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Humans , Integrin beta Chains/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Oligopeptides/pharmacology , Phenethylamines , Propylamines , Protein Transport/drug effects , Rats , Receptors, Calcium-Sensing/chemistry , Signal Transduction/drug effects
9.
J Neurochem ; 120(4): 552-63, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22145864

ABSTRACT

The regulation of pre-synaptic glutamate release is important in the maintenance and fidelity of excitatory transmission in the nervous system. In this study, we report a novel interaction between a ligand-gated ion channel and a G-protein coupled receptor which regulates glutamate release from parallel fiber axon terminals. Immunocytochemical analysis revealed that GABA(A) receptors and the high affinity group III metabotropic glutamate receptor subtype 4 (mGlu4) are co-localized on glutamatergic parallel fiber axon terminals in the cerebellum. GABA(A) and mGlu4 receptors were also found to co-immunoprecipitate from cerebellar membranes. Independently, these two receptors have opposing roles on glutamate release: pre-synaptic GABA(A) receptors promote, while mGlu4 receptors inhibit, glutamate release. However, coincident activation of GABA(A) receptors with muscimol and mGlu4 with the agonist (2S)-S-2-amino-4-phosphonobutanoic acid , increased glutamate release from [(3) H]glutamate-loaded cerebellar synaptosomes above that observed with muscimol alone. Further support for an interaction between GABA(A) and mGlu4 receptors was obtained in the mGlu4 knockout mouse which displayed reduced binding of the GABA(A) ligand [(35) S]tert-butylbicyclophosphorothionate, and decreased expression of the α1, α6, ß2 GABA(A) receptor subunits in the cerebellum. Taken together, our data suggest a new role for mGlu4 whereby simultaneous activation with GABA(A) receptors acts to amplify glutamate release at parallel fiber-Purkinje cell synapses.


Subject(s)
Cerebellum/metabolism , Glutamic Acid/metabolism , Presynaptic Terminals/metabolism , Receptors, GABA-A/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Cerebellum/physiology , GABA Antagonists/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Presynaptic Terminals/physiology , Rats , Receptors, GABA-A/metabolism , Receptors, Presynaptic/physiology
10.
Mol Cell Neurosci ; 46(3): 563-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21215802

ABSTRACT

Fragile X syndrome (FXS), the most common cause of inherited mental retardation, is caused by the loss of the mRNA binding protein, FMRP. Persons with FXS also display epileptic seizures, social anxiety, hyperactivity, and autistic behaviors. The metabotropic glutamate receptor theory of FXS postulates that in the absence of FMRP, enhanced signaling though G-protein coupled group I metabotropic glutamate receptors in the brain contributes to many of the abnormalities observed in the disorder. However, recent evidence suggests that alterations in cellular signaling through additional G-protein coupled receptors may also be involved in the pathogenesis of FXS, thus providing impetus for examining downstream molecules. One group of signaling molecules situated downstream of the receptors is the regulator of G-protein signaling (RGS) proteins. Notably, RGS4 is highly expressed in brain and has been shown to negatively regulate signaling through Group I mGluRs and GABA(B) receptors. To examine the potential role for RGS4 in the pathogenesis of FXS, we generated FXS/RGS4 double knockout mice. Characterization of these mice revealed that a subset of FXS related phenotypes, including increased body weight, altered synaptic protein expression, and abnormal social behaviors, were rescued in the double knockout mice. Other phenotypes, such as hyperactivity and macroorchidism, were not affected by the loss of RGS4. These findings suggest that tissue and cell-type specific differences in GPCR signaling and RGS function may contribute to the spectrum of phenotypic differences observed in FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome , Gene Deletion , Phenotype , RGS Proteins/genetics , Animals , Behavior, Animal/physiology , Body Weight , Disks Large Homolog 4 Protein , Female , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Guanylate Kinases , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Organ Size , RGS Proteins/metabolism , Receptors, GABA-A/metabolism , Signal Transduction/physiology , Social Behavior , Testis/anatomy & histology
11.
J Pharmacol Exp Ther ; 338(3): 897-905, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21636656

ABSTRACT

The most common cause of inherited mental retardation, fragile X syndrome, results from a triplet repeat expansion in the FMR1 gene and loss of the mRNA binding protein, fragile X mental retardation protein (FMRP). In the absence of FMRP, signaling through group I metabotropic glutamate receptors (mGluRs) is enhanced. We previously proposed a mechanism whereby the audiogenic seizures exhibited by FMR1 null mice result from an imbalance in excitatory mGluR and inhibitory GABA(B) receptor (GABA(B)R) signaling (Mol Pharmacol 76:18-24, 2009). Here, we tested the mGluR5-positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB), the mGluR5 inverse agonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), and GABA(B) receptor agonists, alone and in combination on receptor protein expression and audiogenic seizures in FMR1 mice. Single doses of MPEP (30 mg/kg), the GABA(B)R orthosteric agonist R-baclofen (1 mg/kg), or the GABA(B)R-positive allosteric modulator N,N'-dicyclopentyl-2-(methylthio)-5-nitro-4,6-pyrimidine diamine (GS-39783) (30 mg/kg), reduced the incidence of seizures. However, when administered subchronically (daily injections for 6 days), MPEP retained its anticonvulsant activity, whereas R-baclofen and GS-39783 did not. When administered at lower doses that had no effect when given alone, a single injection of MPEP plus R-baclofen also reduced seizures, but the effect was lost after subchronic administration. We were surprised to find that subchronic treatment with R-baclofen also induced tolerance to a single high dose of MPEP. These data demonstrate that tolerance develops rapidly to the antiseizure properties of R-baclofen alone and R-baclofen coadministered with MPEP, but not with MPEP alone. Our findings suggest that cross-talk between the G-protein signaling pathways of these receptors affects drug efficacy after repeated treatment.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/drug therapy , Receptors, GABA-B/drug effects , Receptors, Metabotropic Glutamate/agonists , Animals , Anticonvulsants/pharmacology , Baclofen/administration & dosage , Baclofen/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacology , Blotting, Western , Cyclopentanes/administration & dosage , Cyclopentanes/pharmacology , Drug Interactions , Drug Tolerance , Epilepsy, Reflex/prevention & control , Excitatory Amino Acid Antagonists/pharmacology , GABA Agonists/administration & dosage , GABA Agonists/pharmacology , GABA Modulators/administration & dosage , GABA Modulators/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptors, Kainic Acid/drug effects
12.
Exp Brain Res ; 210(3-4): 451-63, 2011 May.
Article in English | MEDLINE | ID: mdl-21336828

ABSTRACT

This paper reviews results that support a model in which memory for VOR gain is initially encoded in the flocculus, and in which cerebellar LTD and LTP are responsible for gain increases and gain decreases, respectively. We also review data suggesting that after it is encoded, motor memory can either be disrupted, possibly by a local mechanism, or else consolidated. We show that consolidation can be rapid, in which case the frequency dependence of learning is unchanged and we will argue that this is consistent with a local mechanism of consolidation. In the longer term, however, the available evidence supports the transfer of memory out of the flocculus. In new experiments reported here, we address the mechanism of memory encoding. Pharmacological evidence shows that both mGluR1 and GABA(B) receptors in the flocculus are necessary for gain-up, but not for gain-down learning. Immunohistochemical experiments show that the two receptors are largely segregated on different dendritic spines on Purkinje cells. Together with what is already known of the mechanisms of cerebellar LTD and LTP, our data suggest that the direction of learning may be determined by interactions among groups of spines. Our results also provide new evidence for the existence of frequency channels for vestibular signals within the cerebellar cortex.


Subject(s)
Cerebellum/physiology , Learning/physiology , Movement/physiology , Neuronal Plasticity/physiology , Reflex, Vestibulo-Ocular/physiology , Animals , Cerebellum/cytology , Excitatory Amino Acid Agents/pharmacology , GABA Agents/pharmacology , Humans , Learning/drug effects , Memory/physiology , Models, Biological , Neuronal Plasticity/drug effects , Receptors, GABA-A/metabolism , Receptors, Metabotropic Glutamate/metabolism , Reflex, Vestibulo-Ocular/drug effects
13.
Front Neurol ; 12: 745159, 2021.
Article in English | MEDLINE | ID: mdl-34671313

ABSTRACT

Several neurological and psychiatric disorders have been associated with impairments in GABAergic inhibitory neurons in the brain. Thus, in the current era of accelerated development of molecular medicine and biologically-based drugs, there is a need to identify gene regulatory sequences that can be utilized for selectively manipulating the expression of nucleic acids and proteins in GABAergic neurons. This is particularly important for the use of viral vectors in gene therapy. In this Mini Review, we discuss the use of various gene regulatory elements for targeting GABAergic neurons, with an emphasis on adeno-associated viral vectors, the most widely used class of viral vectors for treating brain diseases.

14.
Mol Ther Methods Clin Dev ; 22: 196-209, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34485605

ABSTRACT

Fragile X syndrome (FXS), a neurodevelopmental disorder with no known cure, is caused by a lack of expression of the fragile X mental retardation protein (FMRP). As a single-gene disorder, FXS is an excellent candidate for viral-vector-based gene therapy, although that is complicated by the existence of multiple isoforms of FMRP, whose individual cellular functions are unknown. We studied the effects of rat and mouse orthologs of human isoform 17, a major expressed isoform of FMRP. Injection of neonatal Fmr1 knockout rats and mice with adeno-associated viral vectors (AAV9 serotype) under the control of an MeCP2 mini-promoter resulted in widespread distribution of the FMRP transgenes throughout the telencephalon and diencephalon. Transgene expression occurred mainly in non-GABAergic neurons, with little expression in glia. Early postnatal treatment resulted in partial rescue of the Fmr1 KO rat phenotype, including improved social dominance in treated Fmr1 KO females and partial rescue of locomotor activity in males. Electro-encephalogram (EEG) recordings showed correction of abnormal slow-wave activity during the sleep-like state in male Fmr1 KO rats. These findings support the use of AAV-based gene therapy as a treatment for FXS and specifically demonstrate the potential therapeutic benefit of human FMRP isoform 17 orthologs.

15.
J Neurosci ; 29(25): 8143-55, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19553454

ABSTRACT

Deactivation of glutamatergic signaling in the brain is mediated by glutamate uptake into glia and neurons by glutamate transporters. Glutamate transporters are sodium-dependent proteins that putatively rely indirectly on Na,K-ATPases to generate ion gradients that drive transmitter uptake. Based on anatomical colocalization, mutual sodium dependency, and the inhibitory effects of the Na,K-ATPase inhibitor ouabain on glutamate transporter activity, we postulated that glutamate transporters are directly coupled to Na,K-ATPase and that Na,K-ATPase is an essential modulator of glutamate uptake. Na,K-ATPase was purified from rat cerebellum by tandem anion exchange and ouabain affinity chromatography, and the cohort of associated proteins was characterized by mass spectrometry. The alpha1-alpha 3 subunits of Na,K-ATPase were detected, as were the glutamate transporters GLAST and GLT-1, demonstrating that glutamate transporters copurify with Na,K-ATPases. The link between glutamate transporters and Na,K-ATPase was further established by coimmunoprecipitation and colocalization. Analysis of the regulation of glutamate transporter and Na,K-ATPase activities was assessed using [(3)H]D-aspartate, [(3)H]L-glutamate, and rubidium-86 uptake into synaptosomes and cultured astrocytes. In synaptosomes, ouabain produced a dose-dependent inhibition of glutamate transporter and Na,K-ATPase activities, whereas in astrocytes, ouabain showed a bimodal effect whereby glutamate transporter activity was stimulated at 1 microm ouabain and inhibited at higher concentrations. The effects of protein kinase inhibitors on [(3)H]D-aspartate uptake indicated the selective involvement of Src kinases, which are probably a component of the Na,K-ATPase/glutamate transporter complex. These findings demonstrate that glutamate transporters and Na,K-ATPases are part of the same macromolecular complexes and operate as a functional unit to regulate glutamatergic neurotransmission.


Subject(s)
Astrocytes/metabolism , Cerebellum/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Synaptosomes/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid/metabolism , Blotting, Western , Cell Culture Techniques , Cerebellum/cytology , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Rubidium/metabolism
16.
Neuroimage ; 53(3): 1023-9, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20304074

ABSTRACT

Fragile X Syndrome (FXS) is the most common single gene cause of inherited mental impairment, and cognitive deficits can range from simple learning disabilities to mental retardation. Human FXS is caused by a loss of the Fragile X Mental Retardation Protein (FMRP). The fragile X knockout (FX KO) mouse also shows a loss of FMRP, as well as many of the physical and behavioural characteristics of human FXS. This work aims to characterize the anatomical changes between the FX KO and a corresponding wild type mouse. Significant volume decreases were found in two regions within the deep cerebellar nuclei, namely the nucleus interpositus and the fastigial nucleus, which may be caused by a loss of neurons as indicated by histological analysis. Well-known links between these nuclei and previously established behavioural and physical characteristics of FXS are discussed. The loss of FMRP has a significant effect on these two nuclei, and future studies of FXS should evaluate the biochemical, physiological, and behavioral consequences of alterations in these key nuclei.


Subject(s)
Cerebellar Nuclei/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Animals , Disease Models, Animal , Image Processing, Computer-Assisted , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Tomography, X-Ray Computed
17.
Anal Biochem ; 405(2): 260-2, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20599655

ABSTRACT

L-Serine-O-phosphate (L-SOP), the precursor of l-serine, is an agonist at group III metabotropic glutamate receptors. Despite the interest in L-SOP, very few articles have reported its brain levels. Here we report a convenient and reproducible method for simultaneous analysis of L-SOP and several other important amino acids in brain tissue using high-performance liquid chromatography (HPLC) with fluorimetric detection after derivatization with o-phthaldialdehyde and N-isobutyl-L-cysteine. Analyses were carried out in rat whole brain and cerebellum and in mouse whole brain, forebrain, amygdala, and prefrontal cortex. The method should be useful for future comprehensive neurochemical and pharmacological studies on neuropsychiatric disorders.


Subject(s)
Brain Chemistry , Chromatography, High Pressure Liquid/methods , Fluorometry/methods , Phosphoserine/analysis , Animals , Brain/metabolism , Cerebellum , Mice , Mice, Inbred C57BL , Prefrontal Cortex/chemistry , Rats , Rats, Sprague-Dawley , Serine/metabolism
18.
Hum Gene Ther ; 31(5-6): 339-351, 2020 03.
Article in English | MEDLINE | ID: mdl-31830809

ABSTRACT

Dravet syndrome (DS) is a neurodevelopmental genetic disorder caused by mutations in the SCN1A gene encoding the α subunit of the NaV1.1 voltage-gated sodium channel that controls neuronal action potential firing. The high density of this mutated channel in GABAergic interneurons results in impaired inhibitory neurotransmission and subsequent excessive activation of excitatory neurons. The syndrome is associated with severe childhood epilepsy, autistic behaviors, and sudden unexpected death in epilepsy. Here, we compared the rescue effects of an adeno-associated viral (AAV) vector coding for the multifunctional ß1 sodium channel auxiliary subunit (AAV-NaVß1) with a control vector lacking a transgene. We hypothesized that overexpression of NaVß1 would facilitate the function of residual voltage-gated channels and improve the DS phenotype in the Scn1a+/- mouse model of DS. AAV-NaVß1 was injected into the cerebral spinal fluid of neonatal Scn1a+/- mice. In untreated control Scn1a+/- mice, females showed a higher degree of mortality than males. Compared with Scn1a+/- control mice, AAV-NaVß1-treated Scn1a+/- mice displayed increased survival, an outcome that was more pronounced in females than males. In contrast, behavioral analysis revealed that male, but not female, Scn1a+/- mice displayed motor hyperactivity, and abnormal performance on tests of fear and anxiety and learning and memory. Male Scn1a+/- mice treated with AAV-NaVß1 showed reduced spontaneous seizures and normalization of motor activity and performance on the elevated plus maze test. These findings demonstrate sex differences in mortality in untreated Scn1a+/- mice, an effect that may be related to a lower level of intrinsic inhibitory tone in female mice, and a normalization of aberrant behaviors in males after central nervous system administration of AAV-NaVß1. The therapeutic efficacy of AAV-NaVß1 in a mouse model of DS suggests a potential new long-lasting biological therapeutic avenue for the treatment of this catastrophic epilepsy.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/therapy , Genetic Therapy , NAV1.1 Voltage-Gated Sodium Channel/genetics , Voltage-Gated Sodium Channel beta-1 Subunit/genetics , Animals , Autistic Disorder/genetics , Autistic Disorder/therapy , Dependovirus/genetics , Disease Models, Animal , Epilepsy/genetics , Epilepsy/therapy , Female , Genetic Vectors/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Seizures/genetics , Seizures/therapy , Sex Factors , Transgenes , Treatment Outcome
19.
Mol Pharmacol ; 76(1): 18-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19351745

ABSTRACT

Mice lacking the gene encoding fragile X mental retardation protein (FMR1) are susceptible to audiogenic seizures, and antagonists of the group I metabotropic glutamate receptors (mGluRs) have been shown to block seizures in FMR1 knockout mice. We investigated whether the G-protein-inhibitory activity of the regulator of G-protein signaling protein, RGS4, could also alter the susceptibility to audiogenic seizures in FMR1 mice. We were surprised to find that male FMR1/RGS4 double-knockout mice showed reduced susceptibility to audiogenic seizures compared with age-matched FMR1 mice. These data raised the intriguing possibility that loss of RGS4 increased signaling through another G-protein pathway that reduces seizure susceptibility in FMR1 mice. Indeed, administration of the GABA(B) receptor agonist baclofen to FMR1 mice inhibited seizures, whereas the GABA(B) receptor antagonist (3-aminopropyl)(cyclohexylmethyl)phosphinic acid (CGP 46381) increased seizure incidence in double-knockout mice but not in wild-type mice. Finally, audiogenic seizures could be induced in wild-type mice by coadministering CGP 46381 and the mGluR5-positive allosteric modulator 3-cyano-N-(1,2 diphenyl-1H-pyrazol-5-yl) benzamide. These data show for the first time that GABA(B) receptor-mediated signaling antagonizes the seizure-promoting effects of the mGluRs in FMR1 knockout mice and point to the potential therapeutic benefit of GABA(B) agonists for the treatment of fragile X syndrome.


Subject(s)
Fragile X Mental Retardation Protein/physiology , RGS Proteins/physiology , Receptors, GABA-B/physiology , Seizures/prevention & control , Signal Transduction/physiology , Animals , Baclofen/pharmacology , Disease Susceptibility , Female , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphinic Acids/pharmacology , RGS Proteins/genetics , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/physiology
20.
J Pharmacol Exp Ther ; 330(1): 212-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19389924

ABSTRACT

The peptide N-acetylaspartylglutamate (NAAG) is present in high concentrations in the mammalian central nervous system. Various mechanisms have been proposed for its action, including selective activation of the metabotropic glutamate receptor (mGluR) subtype 3, its action at the N-methyl-D-aspartate receptor, or the production of glutamate by its hydrolysis catalyzed by an extracellular protease. To re-examine its agonist activity at mGluR3, we coexpressed human or rat mGluR3 with G protein inward rectifying channels in Xenopus laevis oocytes. High-performance liquid chromatography analysis of commercial sources of NAAG showed 0.38 to 0.48% glutamate contamination. Although both human and rat mGluR3 were highly sensitive to glutamate, with EC(50) values of 58 and 28 nM, respectively, purified NAAG (100 microM) had little activity (7.7% of full activation by glutamate). Only in the millimolar range did it show significant activity, possibly due to residual traces of glutamate remaining in the purified NAAG preparations. In contrast, the unpurified NAAG sample did produce a full agonist response with mGluR3 coexpressed with G alpha(15), with an EC(50) of 120 microM, as measured by a calcium release assay. This response can be explained by the 0.38 to 0.48% glutamate contamination. Our results suggest that NAAG may not have a direct agonist activity at the mGluR3 receptor. Thus, several in vivo and in vitro published results that did not address the issue of glutamate contamination of NAAG preparations may need to be re-evaluated.


Subject(s)
Dipeptides/agonists , Dipeptides/pharmacology , Neuropeptides/agonists , Neuropeptides/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/physiology , Animals , Cell Line , Female , Humans , Oocytes/metabolism , Rats , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL