Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Ann Neurol ; 94(4): 658-671, 2023 10.
Article in English | MEDLINE | ID: mdl-37243335

ABSTRACT

OBJECTIVE: Spinocerebellar ataxia type 3 (SCA3) is the most common dominantly inherited ataxia, and biomarkers are needed to noninvasively monitor disease progression and treatment response. Anti-ATXN3 antisense oligonucleotide (ASO) treatment has been shown to mitigate neuropathology and rescue motor phenotypes in SCA3 mice. Here, we investigated whether repeated ASO administration reverses brainstem and cerebellar neurochemical abnormalities by magnetic resonance spectroscopy (MRS). METHODS: Symptomatic SCA3 mice received intracerebroventricular treatment of ASO or vehicle and were compared to wild-type vehicle-treated littermates. To quantify neurochemical changes in treated mice, longitudinal 9.4T MRS of cerebellum and brainstem was performed. Acquired magnetic resonance (MR) group means were analyzed by 2-way analysis of variance mixed-effects sex-adjusted analysis with post hoc Sidak correlation for multiple comparisons. Pearson correlations were used to relate SCA3 pathology and behavior. RESULTS: MR spectra yielded 15 to 16 neurochemical concentrations in the cerebellum and brainstem. ASO treatment in SCA3 mice resulted in significant total choline rescue and partial reversals of taurine, glutamine, and total N-acetylaspartate across both regions. Some ASO-rescued neurochemicals correlated with reduction in diseased protein and nuclear ATXN3 accumulation. ASO-corrected motor activity correlated with total choline and total N-acetylaspartate levels early in disease. INTERPRETATION: SCA3 mouse cerebellar and brainstem neurochemical trends parallel those in patients with SCA3. Decreased total choline may reflect oligodendrocyte abnormalities, decreased total N-acetylaspartate highlights neuronal health disturbances, and high glutamine may indicate gliosis. ASO treatment fully or partially reversed select neurochemical abnormalities in SCA3 mice, indicating the potential for these measures to serve as noninvasive treatment biomarkers in future SCA3 gene silencing trials. ANN NEUROL 2023;94:658-671.


Subject(s)
Machado-Joseph Disease , Neurochemistry , Humans , Mice , Animals , Machado-Joseph Disease/genetics , Machado-Joseph Disease/pathology , Oligonucleotides, Antisense/therapeutic use , Glutamine , Biomarkers , Choline/metabolism
2.
Magn Reson Med ; 86(3): 1544-1559, 2021 09.
Article in English | MEDLINE | ID: mdl-33821502

ABSTRACT

PURPOSE: The primary goal of this study was to investigate whether chronic exposures to ultra-high B0 fields can induce long-term cognitive, behavioral, or biological changes in C57BL/6 mice. METHODS: C57BL/6 mice were chronically exposed to 10.5-T or 16.4-T magnetic fields (3-h exposures, two exposure sessions per week, 4 or 8 weeks of exposure). In vivo single-voxel 1 H magnetic resonance spectroscopy was used to investigate possible neurochemical changes in the hippocampus. In addition, a battery of behavioral tests, including the Morris water-maze, balance-beam, rotarod, and fear-conditioning tests, were used to examine long-term changes induced by B0 exposures. RESULTS: Hippocampal neurochemical profile, cognitive, and basic motor functions were not impaired by chronic magnetic field exposures. However, the balance-beam-walking test and the Morris water-maze testing revealed B0 -induced changes in motor coordination and balance. The tight-circling locomotor behavior during Morris water-maze tests was found as the most sensitive factor indexing B0 -induced changes. Long-term behavioral changes were observed days or even weeks subsequent to the last B0 exposure at 16.4 T but not at 10.5 T. Fast motion of mice in and out of the 16.4-T magnet was not sufficient to induce such changes. CONCLUSION: Observed results suggest that the chronic exposure to a magnetic field as high as 16.4 T may result in long-term impairment of the vestibular system in mice. Although observation of mice may not directly translate to humans, nevertheless, they indicate that studies focused on human safety at very high magnetic fields are necessary.


Subject(s)
Conditioning, Psychological , Motor Activity , Animals , Behavior, Animal , Magnetic Fields , Mice , Mice, Inbred C57BL
3.
Pediatr Res ; 89(4): 838-845, 2021 03.
Article in English | MEDLINE | ID: mdl-32555536

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) is a leading infectious cause of neurologic deficits, both in the settings of congenital and perinatal infection, but few animal models exist to study neurodevelopmental outcomes. This study examined the impact of neonatal guinea pig CMV (GPCMV) infection on spatial learning and memory in a Morris water maze (MWM) model. METHODS: Newborn pups were challenged intraperitoneally (i.p.) with a pathogenic red fluorescent protein-tagged GPCMV, or sham inoculated. On days 15-19 post infection (p.i.), pups were tested in the MWM. Viral loads were measured in blood and tissue by quantitative PCR (qPCR), and brain samples collected at necropsy were examined by histology and immunohistochemistry. RESULTS: Viremia (DNAemia) was detected at day 3 p.i. in 7/8 challenged animals. End-organ dissemination was observed, by qPCR, in the lung, liver, and spleen. CD4-positive (CD4+) and CD8-positive (CD8+) T cell infiltrates were present in brains of challenged animals, particularly in periventricular and hippocampal regions. Reactive gliosis and microglial nodules were observed. Statistically significant spatial learning and memory deficits were observed by MWM, particularly for total maze distance traveled (p < 0.0001). CONCLUSION: Neonatal GPCMV infection in guinea pigs results in cognitive defects demonstrable by the MWM. This neonatal guinea pig challenge model can be exploited for studying antiviral interventions. IMPACT: CMV impairs neonatal neurocognition and memory in the setting of postnatal infection. The MWM can be used to examine memory and learning in a guinea pig model of neonatal CMV infection. CD4+ and CD8+ T cells infiltrate the brain following neonatal CMV challenge. This article demonstrates that the MWM can be used to evaluate memory and learning after neonatal GPCMV challenge. The guinea pig can be used to examine central nervous system pathology caused by neonatal CMV infection and this attribute may facilitate the study of vaccines and antivirals.


Subject(s)
Cytomegalovirus/metabolism , Animals , Animals, Newborn , Antibodies, Viral/metabolism , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Fibroblasts/metabolism , Guinea Pigs , Luminescent Proteins/metabolism , Maze Learning , Neurology/methods , Viral Load , Red Fluorescent Protein
4.
Br J Pharmacol ; 177(24): 5658-5676, 2020 12.
Article in English | MEDLINE | ID: mdl-33080056

ABSTRACT

BACKGROUND AND PURPOSE: A significant number of HIV-1 patients on antiretroviral therapy develop HIV-associated neurocognitive disorders (HAND). Evidence indicate that biological sex may regulate HAND pathogenesis, but the mechanisms remain unknown. We investigated synaptic mechanisms associated with sex differences in HAND, using the HIV-1-transgenic 26 (Tg26) mouse model. EXPERIMENTAL APPROACH: Contextual- and cue-dependent memories of male and female Tg26 mice and littermate wild type mice were assessed in a fear conditioning paradigm. Hippocampal electrophysiology, immunohistochemistry, western blot, qRT-PCR and ELISA techniques were used to investigate cellular, synaptic and molecular impairments. KEY RESULTS: Cue-dependent memory was unaltered in male and female Tg26 mice, when compared to wild type mice. Male, but not female, Tg26 mice showed deficits in contextual fear memory. Consistently, only male Tg26 mice showed depressed hippocampal basal synaptic transmission and impaired LTP induction in area CA1. These deficits in male Tg26 mice were independent of hippocampal neuronal loss and microglial activation but were associated with increased HIV-1 long terminal repeat mRNA expression, reduced hippocampal synapsin-1 protein, reduced BDNF mRNA and protein, reduced AMPA glutamate receptor (GluA1) phosphorylation levels and increased glycogen synthase kinase 3 (GSK3) activity. Importantly, selective GSK3 inhibition using 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione increased levels of synapsin-1, BDNF and phosphorylated-GluA1 proteins, restored hippocampal basal synaptic transmission and LTP, and improved contextual fear memory in male Tg26 mice. CONCLUSION AND IMPLICATIONS: Sex-dependent impairments in contextual fear memory and synaptic plasticity in Tg26 mice are associated with increased GSK3 activity. This implicates GSK3 inhibition as a potential therapeutic strategy to improve cognition in HIV-1 patients.


Subject(s)
HIV-1 , Animals , Fear , Female , Glycogen Synthase Kinase 3 , Hippocampus , Humans , Long-Term Potentiation , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Acta Neuropathol ; 138(4): 551-574, 2019 10.
Article in English | MEDLINE | ID: mdl-31168644

ABSTRACT

Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are clinically and neuropathologically highly related α-synucleinopathies that collectively constitute the second leading cause of neurodegenerative dementias. Genetic and neuropathological studies directly implicate α-synuclein (αS) abnormalities in PDD and DLB pathogenesis. However, it is currently unknown how αS abnormalities contribute to memory loss, particularly since forebrain neuronal loss in PDD and DLB is less severe than in Alzheimer's disease. Previously, we found that familial Parkinson's disease-linked human mutant A53T αS causes aberrant localization of the microtubule-associated protein tau to postsynaptic spines in neurons, leading to postsynaptic deficits. Thus, we directly tested if the synaptic and memory deficits in a mouse model of α-synucleinopathy (TgA53T) are mediated by tau. TgA53T mice exhibit progressive memory deficits associated with postsynaptic deficits in the absence of obvious neuropathological and neurodegenerative changes in the hippocampus. Significantly, removal of endogenous mouse tau expression in TgA53T mice (TgA53T/mTau-/-), achieved by mating TgA53T mice to mouse tau-knockout mice, completely ameliorates cognitive dysfunction and concurrent synaptic deficits without affecting αS expression or accumulation of selected toxic αS oligomers. Among the known tau-dependent effects, memory deficits in TgA53T mice were associated with hippocampal circuit remodeling linked to chronic network hyperexcitability. This remodeling was absent in TgA53T/mTau-/- mice, indicating that postsynaptic deficits, aberrant network hyperactivity, and memory deficits are mechanistically linked. Our results directly implicate tau as a mediator of specific human mutant A53T αS-mediated abnormalities related to deficits in hippocampal neurotransmission and suggest a mechanism for memory impairment that occurs as a consequence of synaptic dysfunction rather than synaptic or neuronal loss. We hypothesize that these initial synaptic deficits contribute to network hyperexcitability which, in turn, exacerbate cognitive dysfunction. Our results indicate that these synaptic changes present potential therapeutic targets for amelioration of memory deficits in α-synucleinopathies.


Subject(s)
Memory Disorders/metabolism , Synapses/metabolism , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Transgenic , Neuronal Plasticity , Neurons/metabolism , Neurons/pathology , Synapses/pathology , Synucleinopathies/genetics , Synucleinopathies/pathology , alpha-Synuclein/genetics , tau Proteins/genetics
6.
J Neurosci ; 39(24): 4785-4796, 2019 06 12.
Article in English | MEDLINE | ID: mdl-30948476

ABSTRACT

Understanding the neurobiological processes that incite drug craving and drive relapse has the potential to help target efforts to treat addiction. The NAc serves as a critical substrate for reward and motivated behavior, in part due to alterations in excitatory synaptic strength within cortical-accumbens pathways. The present studies investigated a causal link between cocaine-induced reinstatement of conditioned place preference and rapid reductions of cocaine-dependent increases in NAc shell synaptic strength in male mice. Cocaine-conditioned place preference behavior and ex vivo whole-cell electrophysiology showed that cocaine-primed reinstatement and synaptic depotentiation were disrupted by inhibiting AMPAR internalization via intra-NAc shell infusion of a Tat-GluA23Y peptide. Furthermore, reinstatement was driven by an mGluR5-dependent reduction in AMPAR signaling. Intra-NAc shell infusion of the mGluR5 antagonist MTEP blocked cocaine-primed reinstatement and corresponding depotentiation, whereas infusion of the mGluR5 agonist CHPG itself promoted reinstatement and depotentiated synaptic strength in the NAc shell. Optogenetic examination of circuit-specific plasticity showed that inhibition of infralimbic cortical input to the NAc shell blocked cocaine-primed reinstatement, whereas low-frequency stimulation (10 Hz) of this pathway in the absence of cocaine triggered a reduction in synaptic strength akin to that observed with cocaine, and was sufficient to promote reinstatement in the absence of a cocaine challenge. These data support a model in which mGluR5-mediated reduction in GluA2-containing AMPARs at NAc shell synapses receiving input from the infralimbic cortex is a critical factor in triggering reinstatement of cocaine-primed conditioned approach behavior.SIGNIFICANCE STATEMENT These studies identified a sequence of neural events whereby reexposure to cocaine activates a signaling cascade that alters synaptic strength in the NAc shell and triggers a behavioral response driven by a drug-associated memory.


Subject(s)
Cocaine/pharmacology , Conditioning, Operant/drug effects , Nucleus Accumbens/metabolism , Receptors, Kainic Acid/metabolism , Synaptic Potentials/drug effects , Animals , Cocaine/antagonists & inhibitors , Electrophysiological Phenomena , Long-Term Synaptic Depression/drug effects , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Optogenetics , Patch-Clamp Techniques , Piperidines/pharmacology , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Thiazoles/pharmacology
7.
Br J Pharmacol ; 176(13): 2238-2249, 2019 07.
Article in English | MEDLINE | ID: mdl-30924523

ABSTRACT

BACKGROUND AND PURPOSE: G protein-gated inwardly rectifying K+ (Kir 3) channels moderate the activity of excitable cells and have been implicated in neurological disorders and cardiac arrhythmias. Most neuronal Kir 3 channels consist of Kir 3.1 and Kir 3.2 subtypes, while cardiac Kir 3 channels consist of Kir 3.1 and Kir 3.4 subtypes. Previously, we identified a family of urea-containing Kir 3 channel activators, but these molecules exhibit suboptimal pharmacokinetic properties and modest selectivity for Kir 3.1/3.2 relative to Kir 3.1/3.4 channels. Here, we characterize a non-urea activator, VU0810464, which displays nanomolar potency as a Kir 3.1/3.2 activator, improved selectivity for neuronal Kir 3 channels, and improved brain penetration. EXPERIMENTAL APPROACH: We used whole-cell electrophysiology to measure the efficacy and potency of VU0810464 in neurons and the selectivity of VU0810464 for neuronal and cardiac Kir 3 channel subtypes. We tested VU0810464 in vivo in stress-induced hyperthermia and elevated plus maze paradigms. Parallel studies with ML297, the prototypical activator of Kir 3.1-containing Kir 3 channels, were performed to permit direct comparisons. KEY RESULTS: VU0810464 and ML297 exhibited comparable efficacy and potency as neuronal Kir 3 channel activators, but VU0810464 was more selective for neuronal Kir 3 channels. VU0810464, like ML297, reduced stress-induced hyperthermia in a Kir 3-dependent manner in mice. ML297, but not VU0810464, decreased anxiety-related behaviour as assessed with the elevated plus maze test. CONCLUSION AND IMPLICATIONS: VU0810464 represents a new class of Kir 3 channel activator with enhanced selectivity for Kir 3.1/3.2 channels. VU0810464 may be useful for examining Kir 3.1/3.2 channel contributions to complex behaviours and for probing the potential of Kir 3 channel-dependent manipulations to treat neurological disorders.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Neurons/drug effects , Animals , Anxiety/physiopathology , Behavior, Animal/drug effects , Brain/cytology , Brain/metabolism , Cells, Cultured , Female , Fever/etiology , Fever/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Sinoatrial Node/cytology , Stress, Psychological/complications , Stress, Psychological/physiopathology
8.
J Neurosci ; 38(45): 9754-9767, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30249789

ABSTRACT

Abnormalities in α-synuclein are implicated in the pathogenesis of Parkinson's disease (PD). Because α-synuclein is highly concentrated within presynaptic terminals, presynaptic dysfunction has been proposed as a potential pathogenic mechanism. Here, we report novel, tau-dependent, postsynaptic deficits caused by A53T mutant α-synuclein, which is linked to familial PD. We analyzed synaptic activity in hippocampal slices and cultured hippocampal neurons from transgenic mice of either sex expressing human WT, A53T, and A30P α-synuclein. Increased α-synuclein expression leads to decreased spontaneous synaptic vesicle release regardless of genotype. However, only those neurons expressing A53T α-synuclein exhibit postsynaptic dysfunction, including decreased miniature postsynaptic current amplitude and decreased AMPA to NMDA receptor current ratio. We also found that long-term potentiation and spatial learning were impaired by A53T α-synuclein expression. Mechanistically, postsynaptic dysfunction requires glycogen synthase kinase 3ß-mediated tau phosphorylation, tau mislocalization to dendritic spines, and calcineurin-dependent AMPA receptor internalization. Previous studies reveal that human A53T α-synuclein has a high aggregation potential, which may explain the mutation's unique capacity to induce postsynaptic deficits. However, patients with sporadic PD with severe tau pathology are also more likely to have early onset cognitive decline. Our results here show a novel, functional role for tau: mediating the effects of α-synuclein on postsynaptic signaling. Therefore, the unraveled tau-mediated signaling cascade may contribute to the pathogenesis of dementia in A53T α-synuclein-linked familial PD cases, as well as some subgroups of PD cases with extensive tau pathology.SIGNIFICANCE STATEMENT Here, we report mutation-specific postsynaptic deficits that are caused by A53T mutant α-synuclein, which is linked to familial Parkinson's disease (PD). The overexpression of WT, A53T, or A30P human α-synuclein leads to decreased spontaneous synaptic vesicle release. However, only those neurons expressing A53T α-synuclein exhibit tau phosphorylation-dependent postsynaptic dysfunction, which is characterized by decreased miniature postsynaptic current amplitude and decreased AMPA to NMDA receptor current ratio. The mutation-specific postsynaptic effects caused by human A53T α-synuclein will help us better understand the neurobiological basis of this specific form of familial PD. The differential effects of exogenous human WT, A53T, A30P, and E46K α-synuclein on glutamatergic synaptic responses will help to explain the clinical heterogeneity of sporadic and familial PD.


Subject(s)
Mutation/genetics , Neurodegenerative Diseases/genetics , Synaptic Potentials/physiology , alpha-Synuclein/genetics , tau Proteins/genetics , Animals , Animals, Newborn , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiopathology , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/physiopathology , Organ Culture Techniques , Rats , alpha-Synuclein/biosynthesis , tau Proteins/biosynthesis
9.
Nat Neurosci ; 20(11): 1540-1548, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945222

ABSTRACT

The amygdala plays key roles in fear and anxiety. Studies of the amygdala have largely focused on neuronal function and connectivity. Astrocytes functionally interact with neurons, but their role in the amygdala remains largely unknown. We show that astrocytes in the medial subdivision of the central amygdala (CeM) determine the synaptic and behavioral outputs of amygdala circuits. To investigate the role of astrocytes in amygdala-related behavior and identify the underlying synaptic mechanisms, we used exogenous or endogenous signaling to selectively activate CeM astrocytes. Astrocytes depressed excitatory synapses from basolateral amygdala via A1 adenosine receptor activation and enhanced inhibitory synapses from the lateral subdivision of the central amygdala via A2A receptor activation. Furthermore, astrocytic activation decreased the firing rate of CeM neurons and reduced fear expression in a fear-conditioning paradigm. Therefore, we conclude that astrocyte activity determines fear responses by selectively regulating specific synapses, which indicates that animal behavior results from the coordinated activity of neurons and astrocytes.


Subject(s)
Amygdala/physiology , Astrocytes/physiology , Fear/physiology , Maze Learning/physiology , Nerve Net/physiology , Synapses/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Amygdala/cytology , Amygdala/drug effects , Animals , Astrocytes/drug effects , Fear/drug effects , Fear/psychology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/cytology , Nerve Net/drug effects , Organ Culture Techniques , Receptor, Adenosine A2A/physiology , Synapses/drug effects
10.
Biol Psychiatry ; 80(10): 796-806, 2016 11 15.
Article in English | MEDLINE | ID: mdl-26612516

ABSTRACT

BACKGROUND: Cognitive dysfunction occurs in many debilitating conditions including Alzheimer's disease, Down syndrome, schizophrenia, and mood disorders. The dorsal hippocampus is a critical locus of cognitive processes linked to spatial and contextual learning. G protein-gated inwardly rectifying potassium ion (GIRK/Kir3) channels, which mediate the postsynaptic inhibitory effect of many neurotransmitters, have been implicated in hippocampal-dependent cognition. Available evidence, however, derives primarily from constitutive gain-of-function models that lack cellular specificity. METHODS: We used constitutive and neuron-specific gene ablation models targeting an integral subunit of neuronal GIRK channels (GIRK2) to probe the impact of GIRK channels on associative learning and memory. RESULTS: Constitutive Girk2-/- mice exhibited a striking deficit in hippocampal-dependent (contextual) and hippocampal-independent (cue) fear conditioning. Mice lacking GIRK2 in gamma-aminobutyric acid neurons (GAD-Cre:Girk2flox/flox mice) exhibited a clear deficit in GIRK-dependent signaling in dorsal hippocampal gamma-aminobutyric acid neurons but no evident behavioral phenotype. Mice lacking GIRK2 in forebrain pyramidal neurons (CaMKII-Cre(+):Girk2flox/flox mice) exhibited diminished GIRK-dependent signaling in dorsal, but not ventral, hippocampal pyramidal neurons. CaMKII-Cre(+):Girk2flox/flox mice also displayed a selective impairment in contextual fear conditioning, as both cue fear and spatial learning were intact in these mice. Finally, loss of GIRK2 in forebrain pyramidal neurons correlated with enhanced long-term depression and blunted depotentiation of long-term potentiation at the Schaffer collateral/cornu ammonis 1 synapse in the dorsal hippocampus. CONCLUSIONS: Our data suggest that GIRK channels in dorsal hippocampal pyramidal neurons are necessary for normal learning involving aversive stimuli and support the contention that dysregulation of GIRK-dependent signaling may underlie cognitive dysfunction in some disorders.


Subject(s)
Cognitive Dysfunction/metabolism , Fear/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Hippocampus/metabolism , Learning/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism , Signal Transduction/physiology , Animals , Cognitive Dysfunction/physiopathology , Conditioning, Psychological , Hippocampus/physiopathology , Mice , Mice, Transgenic
11.
J Med Chem ; 57(15): 6383-92, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24978316

ABSTRACT

It is now generally recognized that upon activation by an agonist, ß-arrestin associates with G protein-coupled receptors and acts as a scaffold in creating a diverse signaling network that could lead to adverse effects. As an approach to reducing side effects associated with κ opioid agonists, a series of ß-naltrexamides 3-10 was synthesized in an effort to selectively target putative κ opioid heteromers without recruiting ß-arrestin upon activation. The most potent derivative 3 (INTA) strongly activated KOR-DOR and KOR-MOR heteromers in HEK293 cells. In vivo studies revealed 3 to produce potent antinociception, which, when taken together with antagonism data, was consistent with the activation of both heteromers. 3 was devoid of tolerance, dependence, and showed no aversive effect in the conditioned place preference assay. As immunofluorescence studies indicated no recruitment of ß-arrestin2 to membranes in coexpressed KOR-DOR cells, this study suggests that targeting of specific putative heteromers has the potential to identify leads for analgesics devoid of adverse effects.


Subject(s)
Analgesics/chemistry , Indoles/chemistry , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Analgesics/adverse effects , Analgesics/pharmacology , Animals , Arrestins/metabolism , Avoidance Learning/drug effects , Calcium/metabolism , Drug Tolerance , HEK293 Cells , Humans , Indoles/adverse effects , Indoles/pharmacology , Mice , Naltrexone/adverse effects , Naltrexone/chemistry , Naltrexone/pharmacology , Protein Multimerization , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Stereoisomerism , Structure-Activity Relationship , Substance-Related Disorders/etiology , beta-Arrestins
12.
Proc Natl Acad Sci U S A ; 111(29): 10755-60, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002517

ABSTRACT

ML297 was recently identified as a potent and selective small molecule agonist of G-protein-gated inwardly rectifying K(+) (GIRK/Kir3) channels. Here, we show ML297 selectively activates recombinant neuronal GIRK channels containing the GIRK1 subunit in a manner that requires phosphatidylinositol-4,5-bisphosphate (PIP2), but is otherwise distinct from receptor-induced, G-protein-dependent channel activation. Two amino acids unique to the pore helix (F137) and second membrane-spanning (D173) domain of GIRK1 were identified as necessary and sufficient for the selective activation of GIRK channels by ML297. Further investigation into the behavioral effects of ML297 revealed that in addition to its known antiseizure efficacy, ML297 decreases anxiety-related behavior without sedative or addictive liabilities. Importantly, the anxiolytic effect of ML297 was lost in mice lacking GIRK1. Thus, activation of GIRK1-containing channels by ML297 or derivatives may represent a new approach to the treatment of seizure and/or anxiety disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/drug effects , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology , Amino Acid Sequence , Animals , Baclofen/pharmacology , Behavior, Animal/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism
13.
Cell Mol Neurobiol ; 34(3): 419-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24436034

ABSTRACT

D-Serine, a co-agonist at the NMDA receptor (NMDAR), is synthesized from L-serine by the enzyme serine racemase (SR), which is heavily expressed in the forebrain. Although SR was originally reported to be localized exclusively to astrocytes, recent conditional knock out results demonstrate that little SR is expressed in forebrain astrocytes. As a consequence, the cellular location of its product, D-serine, in the brain is also uncertain. Immunocytochemistry now indicates that SR is expressed primarily in forebrain glutamatergic neurons with the remainder in GABAergic interneurons. We utilized SR deficient (SR-/-) mice, which have <15 % of normal D-serine levels, to validate and optimize a D-serine immunohistochemical method. Nearly all of the D-serine in neocortex and hippocampus (HP) is found in neurons, with virtually no D-serine co-localizing with two astrocyte markers. Interestingly, only a subset of the D-serine positive neurons contained SR in the neocortex and HP. Greater than half of the D-serine positive neurons were GABAergic interneurons, with a majority of these neurons containing parvalbumin and/or somatostatin. Only ~25-40 % of interneurons expressed SR in the neocortex and HP. Finally, we demonstrate in human post-mortem neocortex that SR is found in both excitatory and inhibitory neurons, but not in S100ß-containing astrocytes. In sum, these findings conclusively demonstrate that the majority of D-serine is both synthesized and stored in neurons. It will be important to determine the functional significance for the separation of synthesis and storage of D-serine in neurons, as well as the presence of this NMDAR co-agonist in GABAergic interneurons.


Subject(s)
Neurons/chemistry , Prosencephalon/chemistry , Racemases and Epimerases/analysis , Serine/analysis , Age Factors , Aged , Animals , Humans , Male , Mice , Mice, Knockout , Middle Aged , Neurons/enzymology , Neurons/pathology , Prosencephalon/enzymology , Prosencephalon/pathology , Racemases and Epimerases/biosynthesis , Serine/biosynthesis
14.
Proc Natl Acad Sci U S A ; 110(26): E2400-9, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23729812

ABSTRACT

Schizophrenia is characterized by reduced hippocampal volume, decreased dendritic spine density, altered neuroplasticity signaling pathways, and cognitive deficits associated with impaired hippocampal function. We sought to determine whether this diverse pathology could be linked to NMDA receptor (NMDAR) hypofunction, and thus used the serine racemase-null mutant mouse (SR(-/-)), which has less than 10% of normal brain D-serine, an NMDAR coagonist. We found that D-serine was necessary for the maintenance of long-term potentiation in the adult hippocampal dentate gyrus and for full NMDAR activity on granule cells. SR(-/-) mice had reduced dendritic spines and hippocampal volume. These morphological changes were paralleled by diminished BDNF/Akt/mammalian target of rapamycin (mTOR) signaling and impaired performance on a trace-conditioning memory task. Chronic D-serine treatment normalized the electrophysiological, neurochemical, and cognitive deficits in SR(-/-) mice. These results demonstrate that NMDAR hypofunction can reproduce the numerous hippocampal deficits associated with schizophrenia, which can be reversed by chronic peripheral D-serine treatment.


Subject(s)
Racemases and Epimerases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/etiology , Schizophrenia/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Excitatory Postsynaptic Potentials , Humans , Long-Term Potentiation , Male , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Neuronal Plasticity/drug effects , Racemases and Epimerases/deficiency , Racemases and Epimerases/genetics , Receptor, trkB/metabolism , Risk Factors , Schizophrenia/drug therapy , Serine/metabolism , Serine/therapeutic use , Signal Transduction
15.
Neuropsychopharmacology ; 37(11): 2496-504, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22763616

ABSTRACT

Repeated intermittent exposure to amphetamine (AMPH) results in the development of persistent behavioral and neurological changes. When drug exposure is paired with a specific environment, contextual cues can control conditioned responses, context-specific sensitization, and alterations in dendritic morphology in the nucleus accumbens (NAc). Intact N-methyl-D-aspartate (NMDA) glutamate receptor signaling is thought to be required for associative learning. The acquisition of context-specific behavioral sensitization to AMPH and extinction of conditioned hyperactivity have been investigated in two genetically modified mouse strains: the serine racemase homozygous knockout (SR-/-) and glycine transporter 1 heterozygous mutant (GlyT1-/+). These strains have reciprocally altered NMDA receptor co-agonists, D-serine and glycine, levels that result in decreased (SR-/-) or increased (GlyT1-/+) NMDA receptor signaling. AMPH-induced changes in dendritic morphology in the NAc were also examined. SR-/- mice showed reduced expression of context-specific sensitization and conditioned hyperactivity. However, the conditioned hyperactivity in these mice is completely resistant to extinction. Extinction reversed AMPH-induced increased in NAc spine density in wild-type but not SR-/- mice. GlyT1 -/+ mice showed a more rapid acquisition of sensitization, but no alteration in the extinction of conditioned hyperactivity. The SR-/- data demonstrate that a genetic model of NMDA receptor hypofunction displays a reduced ability to extinguish conditioned responses to drug-associated stimuli. Findings also demonstrate that the morphological changes in the NAc encode conditioned responses that are sensitive to extinction and reduced NMDA receptor activity. NMDA receptor hypofunction may contribute to the comorbidity of substance abuse in schizophrenia.


Subject(s)
Amphetamine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Mutation/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Analysis of Variance , Animals , Behavioral Symptoms/chemically induced , Behavioral Symptoms/genetics , Behavioral Symptoms/physiopathology , Cues , Dendritic Spines/drug effects , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Dose-Response Relationship, Drug , Glycine Plasma Membrane Transport Proteins/genetics , Hyperkinesis/chemically induced , Hyperkinesis/genetics , Hyperkinesis/physiopathology , L-Serine Dehydratase/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neurons/drug effects , Neurons/ultrastructure , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Silver Staining , Time Factors
16.
Cell Mol Neurobiol ; 32(4): 613-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22362148

ABSTRACT

D-serine, which is synthesized by the enzyme serine racemase (SR), is a co-agonist at the N-methyl-D-aspartate receptor (NMDAR). Crucial to an understanding of the signaling functions of D-serine is defining the sites responsible for its synthesis and release. In order to quantify the contributions of astrocytes and neurons to SR and D-serine localization, we used recombinant DNA techniques to effect cell type selective suppression of SR expression in astrocytes (aSRCKO) and in forebrain glutamatergic neurons (nSRCKO). The majority of SR is expressed in neurons: SR expression was reduced by ~65% in nSRCKO cerebral cortex and hippocampus, but only ~15% in aSRCKO as quantified by western blots. In contrast, nSRCKO is associated with only modest decreases in D-serine levels as quantified by HPLC, whereas D-serine levels were unaffected in aSRCKO mice. Liver expression of SR was increased by 35% in the nSRCKO, suggesting a role for peripheral SR in the maintenance of brain D-serine. Electrophysiologic studies of long-term potentiation (LTP) at the Schaffer collateral-CA1 pyramidal neuron synapse revealed no alterations in the aSRCKO mice versus wild-type. LTP induced by a single tetanic stimulus was reduced by nearly 70% in the nSRCKO mice. Furthermore, the mini-excitatory post-synaptic currents mediated by NMDA receptors but not by AMPA receptors were significantly reduced in nSRCKO mice. Our findings indicate that in forebrain, where D-serine appears to be the endogenous co-agonist at NMDA receptors, SR is predominantly expressed in glutamatergic neurons, and co-release of glutamate and D-serine is required for optimal activation of post-synaptic NMDA receptors.


Subject(s)
Cerebral Cortex/enzymology , Glutamic Acid/physiology , Neurons/enzymology , Racemases and Epimerases/deficiency , Racemases and Epimerases/genetics , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Racemases and Epimerases/metabolism
17.
Psychopharmacology (Berl) ; 213(1): 143-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20865248

ABSTRACT

RATIONALE: Enhancement of N-methyl-D: -aspartate receptor (NMDAR) activity through its glycine modulatory site (GMS) is a novel therapeutic approach in schizophrenia. Brain concentrations of endogenous GMS agonist D: -serine and antagonist N-acetyl-aspartylglutamate are regulated by serine racemase (SR) and glutamic acid decarboxylase 2 (GCP2), respectively. Using mice genetically, under-expressing these enzymes may clarify the role of NMDAR-mediated neurotransmission in schizophrenia. OBJECTIVES: We investigated the behavioral effects of two psychotomimetic drugs, the noncompetitive NMDAR antagonist, phencyclidine (PCP; 0, 1.0, 3.0, or 6.0 mg/kg), and the indirect dopamine receptor agonist, amphetamine (AMPH; 0, 1.0, 2.0, or 4.0 mg/kg), in SR -/- and GCP2 -/+ mice. Outcome measures were locomotor activity and prepulse inhibition (PPI) of the acoustic startle reflex. Acute effects of an exogenous GMS antagonist, gavestinel (0, 3.0, or 10.0 mg/kg), on PCP-induced behaviors were examined in wild-type mice for comparison to the mutants with reduced GMS activity. RESULTS: PCP-induced hyperactivity was increased in GCP2 -/+ mice, and PCP-enhanced startle reactivity was increased in SR -/- mice. PCP disruption of PPI was unaffected in either mutant. In contrast, gavestinel attenuated PCP-induced PPI disruption without effect on baseline PPI or locomotor activity. AMPH effects were similar to controls in both mutant strains. CONCLUSIONS: The results of the PCP experiments demonstrate that convergence of pharmacological and genetic manipulations at NMDARs may confound the predictive validity of these preclinical assays for the effects of GMS activation in schizophrenia. The AMPH data provide additional evidence that hyperdopaminergia in schizophrenia may be distinct from NMDAR hypofunction.


Subject(s)
Brain/physiology , Hallucinogens/pharmacology , Motor Activity/physiology , Neural Inhibition/physiology , Racemases and Epimerases/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Reflex, Startle/physiology , Amphetamine/pharmacology , Animals , Brain/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/physiology , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Neural Inhibition/drug effects , Phencyclidine/pharmacology , Racemases and Epimerases/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reflex, Startle/drug effects
18.
Neurosci Lett ; 488(3): 267-71, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21094213

ABSTRACT

Reduction in cortical presynaptic markers, notably parvalbumin (PV), for the chandelier subtype of inhibitory γ-amino-butyric acid (GABA) interneurons is a highly replicated post-mortem finding in schizophrenia. Evidence from genetic and pharmacological studies implicates hypofunction of N-methyl-d-aspartate receptor (NMDAR)-mediated glutamatergic signaling as a critical component of the pathophysiology of schizophrenia. Serine racemase (SR) produces the endogenous NMDAR co-agonist d-serine, and disruption of the SR gene results in reduced NMDAR signaling. SR null mutant (-/-) mice were used to study the link between NMDAR hypofunction and decreased PV expression, assessed by immunoreactive (IR) cell density in the medial prefrontal cortex and hippocampus and protein levels in brain homogenates from the frontal cortex and hippocampus. Contrary to expectations, SR -/- mice showed modest elevations in PV-IR cell density and no difference in PV expression in brain homogenate. To control for these surprising results, we investigated PV expression in mice and rats following subchronic phencyclidine or ketamine treatments in adulthood. PV expression was not affected by drug these treatment in either species, failing to reproduce previously published findings. Our findings challenge the hypothesis that pathological deficits in PV expression are simply a consequence of NMDAR hypofunction.


Subject(s)
Brain/metabolism , Neurons/metabolism , Parvalbumins/biosynthesis , Receptors, N-Methyl-D-Aspartate/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Blotting, Western , Brain/pathology , Disease Models, Animal , Excitatory Amino Acid Antagonists/toxicity , Glutamate Decarboxylase/biosynthesis , Immunohistochemistry , Ketamine/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Phencyclidine/toxicity , Racemases and Epimerases/genetics , Rats , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Schizophrenia/metabolism , Schizophrenia/physiopathology
19.
Neuropsychopharmacology ; 33(9): 2206-16, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17957214

ABSTRACT

Recent clinical studies in schizophrenic patients show that a selective agonist of group II metabotropic glutamate (mGlu) receptors has robust efficacy in treating positive and negative symptoms. Group II mGlu receptor agonists also modulate the in vivo activity of psychotomimetic drugs, reducing the ability of psychotomimetic hallucinogens to increase glutamatergic transmission. The use of mouse models provides an opportunity to investigate the dynamic action that mGlu2/3 receptors play in regulating the behavioral effects of hallucinogen-induced glutamatergic neurotransmission using genetic as well as pharmacological strategies. The current study sought to characterize the use of the two-lever drug discrimination paradigm in ICR (CD-1) mice, using the hallucinogenic 5-HT2A/2C receptor agonist (-)-2,5-dimethoxy-4-bromoamphetamine [(-)-DOB)] as a stimulus-producing drug. The (-)-DOB discriminative stimulus was dose-dependent, generalized to the hallucinogen lysergic acid diethylamide, and was potently blocked by the 5-HT2A receptor antagonist M100907. However, contrary to our prediction, the hallucinogen-induced discriminative stimulus was not regulated by mGlu2/3 receptors. In a series of follow-up studies using hallucinogen-induced head twitch response and phencyclidine-induced hyperlocomotion, it was additionally discovered that the repeated dosing regimen required for discrimination training attenuated the behavioral effects of the mGlu2/3 receptor agonist LY379268. Furthermore chronic studies, using a 14 day (-)-DOB treatment, confirmed that repeated hallucinogen treatment causes a loss of behavioral activity of mGlu2/3 receptors, likely resulting from persistent activation of mGlu2/3 receptors by a hallucinogen-induced hyperglutamatergic state.


Subject(s)
Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/pharmacology , Hallucinogens/pharmacology , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , DOM 2,5-Dimethoxy-4-Methylamphetamine/pharmacology , Amino Acids/pharmacology , Analysis of Variance , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Conditioning, Operant/drug effects , Discrimination, Psychological/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Exploratory Behavior/drug effects , Fluorobenzenes/pharmacology , Locomotion/drug effects , Lysergic Acid Diethylamide/pharmacology , Male , Mice , Mice, Inbred ICR , Piperidines/pharmacology , Time Factors
20.
Mol Pharmacol ; 72(2): 477-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526600

ABSTRACT

Recent clinical studies reveal that selective agonists of group II metabotropic glutamate (mGlu) receptors have robust efficacy in treating positive and negative symptoms in patients with schizophrenia. Group II mGlu receptor agonists also modulate the in vivo activity of psychotomimetic drugs and reduce the ability of psychotomimetic hallucinogens to increase glutamatergic transmission. Because increased excitation of the medial prefrontal cortex (mPFC) has been implicated in pathophysiology of schizophrenia, the ability of group II mGlu receptor agonists to reduce hallucinogenic drug action in this region is believed to be directly related to their antipsychotic efficacy. A novel class of ligands, termed positive allosteric modulators, has recently been identified, displaying exceptional mGlu2 receptor selectivity. These compounds do not activate mGlu2 receptors directly but potentiate the ability of glutamate and other agonists to activate this receptor. We now report that the mGlu2 receptor-selective positive allosteric modulator biphenyl-indanone A (BINA) modulates excitatory neurotransmission in the mPFC and attenuates the in vivo actions of the hallucinogenic 5-HT(2A/2C) receptor agonist (-)2,5-dimethoxy-4-bromoamphetamine [(-)DOB]. BINA attenuates serotonin-induced increases in spontaneous excitatory postsynaptic currents in the mPFC, mimicking the effect of the mGlu2/3 receptor agonist (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine (DCG-IV). In addition, BINA reduced (-)DOB-induced head twitch behavior and Fos expression in mPFC, effects reversed by pretreatment with the mGlu2/3 receptor antagonist 2S-2-amino-2-(1S,2S-2-carboxycyclopropan-1-yl) -3 - (xanth-9-yl-)propionic acid (LY341495). These data confirm the relevance of excitatory signaling in the mPFC to the behavioral actions of hallucinogens and further support the targeting of mGlu2 receptors as a novel strategy for treating glutamatergic dysfunction in schizophrenia.


Subject(s)
Biphenyl Compounds/pharmacology , DOM 2,5-Dimethoxy-4-Methylamphetamine/analogs & derivatives , Hallucinogens/antagonists & inhibitors , Indans/pharmacology , Psychotic Disorders/drug therapy , Receptors, Metabotropic Glutamate/drug effects , DOM 2,5-Dimethoxy-4-Methylamphetamine/antagonists & inhibitors , Allosteric Regulation , Animals , Excitatory Postsynaptic Potentials/drug effects , Male , Mice , Mice, Inbred ICR , Prefrontal Cortex , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...