Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Commun ; 15(1): 5691, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38971801

ABSTRACT

Cholinergic striatal interneurons (ChIs) express the vesicular glutamate transporter 3 (VGLUT3) which allows them to regulate the striatal network with glutamate and acetylcholine (ACh). In addition, VGLUT3-dependent glutamate increases ACh vesicular stores through vesicular synergy. A missense polymorphism, VGLUT3-p.T8I, was identified in patients with substance use disorders (SUDs) and eating disorders (EDs). A mouse line was generated to understand the neurochemical and behavioral impact of the p.T8I variant. In VGLUT3T8I/T8I male mice, glutamate signaling was unchanged but vesicular synergy and ACh release were blunted. Mutant male mice exhibited a reduced DA release in the dorsomedial striatum but not in the dorsolateral striatum, facilitating habit formation and exacerbating maladaptive use of drug or food. Increasing ACh tone with donepezil reversed the self-starvation phenotype observed in VGLUT3T8I/T8I male mice. Our study suggests that unbalanced dopaminergic transmission in the dorsal striatum could be a common mechanism between SUDs and EDs.


Subject(s)
Corpus Striatum , Dopamine , Animals , Male , Dopamine/metabolism , Mice , Corpus Striatum/metabolism , Humans , Acetylcholine/metabolism , Substance-Related Disorders/metabolism , Substance-Related Disorders/genetics , Signal Transduction/drug effects , Glutamic Acid/metabolism , Interneurons/metabolism , Interneurons/drug effects , Feeding and Eating Disorders/metabolism , Feeding and Eating Disorders/genetics , Feeding and Eating Disorders/physiopathology , Mice, Inbred C57BL , Amino Acid Transport Systems, Acidic/metabolism , Amino Acid Transport Systems, Acidic/genetics , Mutation , Mutation, Missense , Vesicular Acetylcholine Transport Proteins
2.
Cell Rep ; 43(7): 114411, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38944834

ABSTRACT

Exposure to stressors has profound effects on sleep that have been linked to serotonin (5-HT) neurons of the dorsal raphe nucleus (DR). However, the DR also comprises glutamatergic neurons expressing vesicular glutamate transporter type 3 (DRVGLUT3), leading us to examine their role. Cell-type-specific tracing revealed that DRVGLUT3 neurons project to brain areas regulating arousal and stress. We found that chemogenetic activation of DRVGLUT3 neurons mimics stress-induced sleep perturbations. Furthermore, deleting VGLUT3 in the DR attenuated stress-induced sleep perturbations, especially after social defeat stress. In the DR, VGLUT3 is found in subsets of 5-HT and non-5-HT neurons. We observed that both populations are activated by acute stress, including those projecting to the ventral tegmental area. However, deleting VGLUT3 in 5-HT neurons minimally affected sleep regulation. These findings suggest that VGLUT3 expression in the DR drives stress-induced sleep perturbations, possibly involving non-5-HT DRVGLUT3 neurons.


Subject(s)
Dorsal Raphe Nucleus , Neurons , Sleep , Stress, Psychological , Animals , Male , Dorsal Raphe Nucleus/metabolism , Mice , Stress, Psychological/metabolism , Neurons/metabolism , Sleep/physiology , Serotonin/metabolism , Mice, Inbred C57BL , Amino Acid Transport Systems, Acidic/metabolism , Amino Acid Transport Systems, Acidic/genetics
3.
eNeuro ; 10(2)2023 02.
Article in English | MEDLINE | ID: mdl-36720646

ABSTRACT

Fear is an emotional mechanism that helps to cope with potential hazards. However, when fear is generalized, it becomes maladaptive and represents a core symptom of posttraumatic stress disorder (PTSD). Converging lines of research show that dysfunction of glutamatergic neurotransmission is a cardinal feature of trauma and stress related disorders such as PTSD. However, the involvement of glutamatergic co-transmission in fear is less well understood. Glutamate is accumulated into synaptic vesicles by vesicular glutamate transporters (VGLUTs). The atypical subtype, VGLUT3, is responsible for the co-transmission of glutamate with acetylcholine, serotonin, or GABA. To understand the involvement of VGLUT3-dependent co-transmission in aversive memories, we used a Pavlovian fear conditioning paradigm in VGLUT3-/- mice. Our results revealed a higher contextual fear memory in these mice, despite a facilitation of extinction. In addition, the absence of VGLUT3 leads to fear generalization, probably because of a pattern separation deficit. Our study suggests that the VGLUT3 network plays a crucial role in regulating emotional memories. Hence, VGLUT3 is a key player in the processing of aversive memories and therefore a potential therapeutic target in stress-related disorders.


Subject(s)
Fear , Synaptic Transmission , Mice , Animals , Fear/physiology , Vesicular Glutamate Transport Proteins/metabolism , Memory Disorders , Glutamic Acid/metabolism
4.
Neuropsychopharmacology ; 48(7): 1067-1077, 2023 06.
Article in English | MEDLINE | ID: mdl-36302847

ABSTRACT

Delayed upregulation of the neuronal chloride extruder KCC2 underlies the progressive shift in GABA signaling polarity during development. Conversely, KCC2 downregulation is observed in a variety of neurological and psychiatric disorders often associated with cognitive impairment. Reduced KCC2 expression and function in mature networks may disrupt GABA signaling and promote anomalous network activities underlying these disorders. However, the causal link between KCC2 downregulation, altered brain rhythmogenesis, and cognitive function remains elusive. Here, by combining behavioral exploration with in vivo electrophysiology we assessed the impact of chronic KCC2 downregulation in mouse dorsal hippocampus and showed it compromises both spatial and contextual memory. This was associated with altered hippocampal rhythmogenesis and neuronal hyperexcitability, with increased burst firing in CA1 neurons during non-REM sleep. Reducing neuronal excitability with terbinafine, a specific Task-3 leak potassium channel opener, occluded the impairment of contextual memory upon KCC2 knockdown. Our results establish a causal relationship between KCC2 expression and cognitive performance and suggest that non-epileptiform rhythmopathies and neuronal hyperexcitability are central to the deficits caused by KCC2 downregulation in the adult mouse brain.


Subject(s)
Symporters , Animals , Mice , Symporters/metabolism , Hippocampus/metabolism , Neurons/metabolism , Brain/metabolism , gamma-Aminobutyric Acid/metabolism
5.
Front Mol Neurosci ; 15: 991732, 2022.
Article in English | MEDLINE | ID: mdl-36176961

ABSTRACT

Striatal cholinergic interneurons (CINs) use acetylcholine (ACh) and glutamate (Glut) to regulate the striatal network since they express vesicular transporters for ACh (VAChT) and Glut (VGLUT3). However, whether ACh and Glut are released simultaneously and/or independently from cholinergic varicosities is an open question. The answer to that question requires the multichannel detection of vesicular transporters at the level of single synaptic vesicle (SV). Here, we used super-resolution STimulated Emission Depletion microscopy (STED) to characterize and quantify the distribution of VAChT and VGLUT3 in CINs SVs. Nearest-neighbor distances analysis between VAChT and VGLUT3-immunofluorescent spots revealed that 34% of CINs SVs contain both VAChT and VGLUT3. In addition, 40% of SVs expressed only VAChT while 26% of SVs contain only VGLUT3. These results suggest that SVs from CINs have the potential to store simultaneously or independently ACh and/or Glut. Overall, these morphological findings support the notion that CINs varicosities can signal with either ACh or Glut or both with an unexpected level of complexity.

6.
Int J Mol Sci ; 23(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35054976

ABSTRACT

Glutamate is the most abundant excitatory amino acid in the central nervous system. Neurons using glutamate as a neurotransmitter can be characterised by vesicular glutamate transporters (VGLUTs). Among the three subtypes, VGLUT3 is unique, co-localising with other "classical" neurotransmitters, such as the inhibitory GABA. Glutamate, manipulated by VGLUT3, can modulate the packaging as well as the release of other neurotransmitters and serve as a retrograde signal through its release from the somata and dendrites. Its contribution to sensory processes (including seeing, hearing, and mechanosensation) is well characterised. However, its involvement in learning and memory can only be assumed based on its prominent hippocampal presence. Although VGLUT3-expressing neurons are detectable in the hippocampus, most of the hippocampal VGLUT3 positivity can be found on nerve terminals, presumably coming from the median raphe. This hippocampal glutamatergic network plays a pivotal role in several important processes (e.g., learning and memory, emotions, epilepsy, cardiovascular regulation). Indirect information from anatomical studies and KO mice strains suggests the contribution of local VGLUT3-positive hippocampal neurons as well as afferentations in these events. However, further studies making use of more specific tools (e.g., Cre-mice, opto- and chemogenetics) are needed to confirm these assumptions.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/physiology , Pyramidal Cells/metabolism , Vesicular Glutamate Transport Proteins/genetics , Vesicular Glutamate Transport Proteins/metabolism , Animals , Biomarkers , Electrophysiological Phenomena , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice, Knockout , Neurotransmitter Agents/metabolism , Signal Transduction , Synaptic Transmission
7.
Mol Neurobiol ; 57(7): 3118-3142, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32474835

ABSTRACT

Vesicular glutamate transporters (VGLUTs) control quantal size of glutamatergic transmission and have been the center of numerous studies over the past two decades. VGLUTs contain two independent transport modes that facilitate glutamate packaging into synaptic vesicles and phosphate (Pi) ion transport into the synaptic terminal. While a transmembrane proton electrical gradient established by a vacuolar-type ATPase powers vesicular glutamate transport, recent studies indicate that binding sites and flux properties for chloride, potassium, and protons within VGLUTs themselves regulate VGLUT activity as well. These intrinsic ionic binding and flux properties of VGLUTs can therefore be modulated by neurophysiological conditions to affect levels of glutamate available for release from synapses. Despite their extraordinary importance, specific and high-affinity pharmacological compounds that interact with these sites and regulate VGLUT function, distinguish between the various modes of transport, and the different isoforms themselves, are lacking. In this review, we provide an overview of the physiologic sites for VGLUT regulation that could modulate glutamate release in an over-active synapse or in a disease state.


Subject(s)
Glutamic Acid/metabolism , Neurons/metabolism , Synapses/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Animals , Gene Expression Regulation , Humans , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Proteins/genetics
8.
Neuropharmacology ; 164: 107902, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31811873

ABSTRACT

Vesicular glutamate transporters (VGLUT1-3) mediate the uptake of glutamate into synaptic vesicles. VGLUTs are pivotal actors of excitatory transmission and of almost all brain functions. Their implication in various pathologies has been clearly documented. Despite their functional importance, the pharmacology of VGLUTs is limited to a few dyes such as Trypan Blue, Rose Bengal or Brilliant Yellow type. Here, we report the design and evaluation of new potent analogs based on Trypan Blue scaffold. Our best compound, named LSP5-2157, has an EC50 of 50 nM on glutamate vesicular uptake. Using a 3D homology model of VGLUT1 and docking experiments, we determined its putative binding subdomains within vesicular glutamate transporters and validated the structural requirement for VGLUT inhibition. To better estimate the specificity and potency of LSP5-2157, we also investigated its ability to block glutamatergic transmission in autaptic hippocampal cells. Neither glutamate receptors nor GABAergic transmission or transmission machinery were affected by LSP5-2157. Low doses of compound reversibly reduce glutamatergic neurotransmission in hippocampal autpases. LSP5-2157 had a low and depressing effect on synaptic efficacy in hippocampal slice. Furthermore, LSP5-2157 had no effect on NMDA-R- mediated fEPSP but reduce synaptic plasticity induced by 3 trains of 100 Hz. Finally, LSP5-2157 had the capacity to inhibit VGLUT3-dependent auditory synaptic transmission in the guinea pig cochlea. In this model, it abolished the compound action potential of auditory nerve at high concentration showing the limited permeation of LSP5-2157 in an in-vivo model. In summary, the new ligand LSP5-2157, has a high affinity and specificity for VGLUTs and shows some permeability in isolated neuron, tissue preparations or in vivo in the auditory system. These findings broaden the field of VGLUTs inhibitors and open the way to their use to assess glutamatergic functions in vitro and in vivo.


Subject(s)
Vesicular Glutamate Transport Proteins/antagonists & inhibitors , Action Potentials/drug effects , Animals , Cochlea/drug effects , Cochlear Nerve/drug effects , Excitatory Postsynaptic Potentials/drug effects , Guinea Pigs , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Inbred C57BL , Models, Molecular , Neurons/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Vesicular Glutamate Transport Proteins/metabolism
9.
Genes Brain Behav ; 18(3): e12528, 2019 03.
Article in English | MEDLINE | ID: mdl-30324647

ABSTRACT

The atypical vesicular glutamate transporter VGLUT3 is present in subpopulations of GABAergic interneurons in the cortex and the hippocampus, in subgroups of serotoninergic neurons in raphe nuclei, and in cholinergic interneurons in the striatum. C56BL/6N mice that no longer express VGLUT3 (VGLUT3-/- ) display anxiety-associated phenotype, increased spontaneous and cocaine-induced locomotor activity and decreased haloperidol-induced catalepsy. Inbred mouse strains differ markedly in their sensitivity to anxiety and behavioral responses elicited by drugs. The purpose of this study was to investigate strain differences in VGLUT3 expression levels and its potential correlates with anxiety and reward-guided behaviors. Five inbred mouse lines were chosen according to their contrasted anxiety and drugs sensitivity: C57BL/6N, C3H/HeN, DBA/2J, 129/Sv, and BALB/c. VGLUT3 protein expression was measured in different brain areas involved in reward or mood regulation (such as the striatum, the hippocampus, and raphe nuclei) and genetic variations in Slc17a8, the gene encoding for VGLUT3, have been explored. These five inbred mouse strains express very different levels of VGLUT3, which cannot be attributed to the genetic variation of the Slc17a8 locus. Furthermore, mice behavior in the open field, elevated plus maze, spontaneous- and cocaine-induced locomotor was highly heterogeneous and only partially correlated to VGLUT3 levels. These data highlight the fact that one single gene polymorphism could not account for VGLUT3 expression variations, and that region specific VGLUT3 expression level variations might play a key role in the modulation of discrete behaviors.


Subject(s)
Amino Acid Transport Systems, Acidic/genetics , Anxiety/genetics , Cocaine-Related Disorders/genetics , Hyperkinesis/genetics , Amino Acid Transport Systems, Acidic/metabolism , Animals , Anxiety/chemically induced , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cocaine/toxicity , Hyperkinesis/chemically induced , Locomotion , Male , Maze Learning , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA
10.
J Neurochem ; 148(6): 779-795, 2019 03.
Article in English | MEDLINE | ID: mdl-30556914

ABSTRACT

Several subtypes of modulatory neurons co-express vesicular glutamate transporters (VGLUTs) in addition to their cognate vesicular transporters. These neurons are believed to establish new forms of neuronal communication. The atypical VGLUT3 is of particular interest since in the striatum this subtype is found in tonically active cholinergic interneurons (TANs) and in a subset of 5-HT fibers. The striatum plays a major role in psychomotor effects induced by amphetamine. Whether and how VGLUT3-operated glutamate/ACh or glutamate/5HT co-transmissions modulates psychostimulants-induced maladaptive behaviors is still unknown. Here, we investigate the involvement of VGLUT3 and glutamate co-transmission in amphetamine-induced psychomotor effects and stereotypies. Taking advantage of constitutive and cell-type specific VGLUT3-deficient mouse lines, we tackled the hypothesis that VGLUT3 could gate psychomotor effects (locomotor activity and stereotypies) induced by acute or chronic administration of amphetamine. Interestingly, VGLUT3-null mice demonstrated blunted amphetamine-induced stereotypies as well as reduced striatal ∆FosB expression. VGLUT3-positive varicosities within the striatum arise in part from 5HT neurons. We tested the involvement of VGLUT3 deletion in serotoninergic neurons in amphetamine-induced stereotypies. Mice lacking VGLUT3 specifically in 5HT fibers showed no alteration to amphetamine sensitivity. In contrast, specific deletion of VGLUT3 in cholinergic neurons partially phenocopied the effects observed in the constitutive knock-out mice. Our results show that constitutive deletion of VGLUT3 modulates acute and chronic locomotor effects induced by amphetamine. They point to the fact that the expression of VGLUT3 in multiple brain areas is pivotal in gating amphetamine-induced psychomotor adaptations. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Amphetamine/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Locomotion/drug effects , Animals , Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Sci Rep ; 8(1): 938, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343737

ABSTRACT

Synaptic loss, plaques and neurofibrillary tangles are viewed as hallmarks of Alzheimer's disease (AD). This study investigated synaptic markers in neocortical Brodmann area 9 (BA9) samples from 171 subjects with and without AD at different levels of cognitive impairment. The expression levels of vesicular glutamate transporters (VGLUT1&2), glutamate uptake site (EAAT2), post-synaptic density protein of 95 kD (PSD95), vesicular GABA/glycine transporter (VIAAT), somatostatin (som), synaptophysin and choline acetyl transferase (ChAT) were evaluated. VGLUT2 and EAAT2 were unaffected by dementia. The VGLUT1, PSD95, VIAAT, som, ChAT and synaptophysin expression levels significantly decreased as dementia progressed. The maximal decrease varied between 12% (synaptophysin) and 42% (som). VGLUT1 was more strongly correlated with dementia than all of the other markers (polyserial correlation = -0.41). Principal component analysis using these markers was unable to differentiate the CDR groups from one another. Therefore, the status of the major synaptic markers in BA9 does not seem to be linked to the cognitive status of AD patients. The findings of this study suggest that the loss of synaptic markers in BA9 is a late event that is only weakly related to AD dementia.


Subject(s)
Alzheimer Disease/metabolism , Biomarkers/metabolism , Cognition/physiology , Prefrontal Cortex/metabolism , Synapses/metabolism , Aged, 80 and over , Choline O-Acetyltransferase/metabolism , Female , Glutamic Acid/metabolism , Humans , Male , Neurons/metabolism , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
12.
J Neuropathol Exp Neurol ; 76(10): 883-897, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28922852

ABSTRACT

Magnesium sulfate (MgSO4) administration to mothers at risk of preterm delivery is proposed as a neuroprotective strategy against neurological alterations such as cerebral palsy in newborns. However, long-term beneficial or adverse effects of MgSO4 and sex-specific sensitivity remain to be investigated. We conducted behavioral and neurochemical studies of MgSO4 effects in males and females, from the perinatal period to adolescence in a mouse model of cerebral neonatal lesion. The lesion was produced in 5-day-old (P5) pups by ibotenate intracortical injection. MgSO4 (600 mg/kg, i.p.) prior to ibotenate prevented lesion-induced sensorimotor alterations in both sexes at P6 and P7. The lesion increased glutamate level at P10 in the prefrontal cortex, which was prevented by MgSO4 in males. In neonatally lesioned adolescent mice, males exhibited more sequelae than females in motor and cognitive functions. In the perirhinal cortex of adolescent mice, the neonatal lesion induced an increase in vesicular glutamate transporter 1 density in males only, which was negatively correlated with cognitive scores. Long-term sequelae were prevented by neonatal MgSO4 administration. MgSO4 never induced short- or long-term deleterious effect on its own. These results also strongly suggest that sex-specific neuroprotection should be foreseen in preterm infants.


Subject(s)
Brain/metabolism , Calcium Channel Blockers/administration & dosage , Gait Disorders, Neurologic/prevention & control , Magnesium Sulfate/administration & dosage , Neurotoxicity Syndromes/complications , Aging/drug effects , Animals , Animals, Newborn , Brain/drug effects , Brain/pathology , Calcium Channel Blockers/blood , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Female , Functional Laterality , Gait Disorders, Neurologic/etiology , Glutamic Acid/metabolism , Ibotenic Acid/toxicity , Longitudinal Studies , Magnesium Sulfate/blood , Male , Mice , Motor Skills/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Sex Factors , Vesicular Glutamate Transport Protein 1/metabolism , gamma-Aminobutyric Acid/metabolism
13.
Front Cell Neurosci ; 11: 140, 2017.
Article in English | MEDLINE | ID: mdl-28559797

ABSTRACT

Hippocampal interneurons release the inhibitory transmitter GABA to regulate excitation, rhythm generation and synaptic plasticity. A subpopulation of GABAergic basket cells co-expresses the GABA/glycine vesicular transporters (VIAAT) and the atypical type III vesicular glutamate transporter (VGLUT3); therefore, these cells have the ability to signal with both GABA and glutamate. GABAergic transmission by basket cells has been extensively characterized but nothing is known about the functional implications of VGLUT3-dependent glutamate released by these cells. Here, using VGLUT3-null mice we observed that the loss of VGLUT3 results in a metaplastic shift in synaptic plasticity at Shaeffer's collaterals - CA1 synapses and an altered theta oscillation. These changes were paralleled by the loss of a VGLUT3-dependent inhibition of GABAergic current in CA1 pyramidal layer. Therefore presynaptic type III metabotropic could be activated by glutamate released from VGLUT3-positive interneurons. This putative presynaptic heterologous feedback mechanism inhibits local GABAergic tone and regulates the hippocampal neuronal network.

14.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28275711

ABSTRACT

The later stages of long-term potentiation (LTP) in vitro and spatial memory in vivo are believed to depend upon gene transcription. Accordingly, considerable attempts have been made to identify both the mechanisms by which transcription is regulated and indeed the gene products themselves. Previous studies have shown that deletion of one regulator of transcription, the mitogen- and stress-activated kinase 1 (MSK1), causes an impairment of spatial memory. Given the ability of MSK1 to regulate gene expression via the phosphorylation of cAMP response element binding protein (CREB) at serine 133 (S133), MSK1 is a plausible candidate as a prime regulator of transcription underpinning synaptic plasticity and learning and memory. Indeed, prior work has revealed the necessity for MSK1 in homeostatic and experience-dependent synaptic plasticity. However, using a knock-in kinase-dead mouse mutant of MSK1, the current study demonstrates that, while the kinase function of MSK1 is important in regulating the phosphorylation of CREB at S133 and basal synaptic transmission in hippocampal area CA1, it is not required for metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD), two forms of LTP or several forms of spatial learning in the watermaze. These data indicate that other functions of MSK1, such as a structural role for the whole enzyme, may explain previous observations of a role for MSK1 in learning and memory.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/cytology , Long-Term Potentiation/physiology , Memory Disorders/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Synaptic Transmission/physiology , Animals , Cues , Cyclic AMP Response Element-Binding Protein/genetics , Disease Models, Animal , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reaction Time/drug effects , Reaction Time/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Serine/metabolism , Synaptic Transmission/genetics
15.
J Neurosci ; 37(15): 4181-4199, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28314816

ABSTRACT

The atypical vesicular glutamate transporter type 3 (VGLUT3) is expressed by subpopulations of neurons using acetylcholine, GABA, or serotonin as neurotransmitters. In addition, VGLUT3 is expressed in the inner hair cells of the auditory system. A mutation (p.A211V) in the gene that encodes VGLUT3 is responsible for progressive deafness in two unrelated families. In this study, we investigated the consequences of the p.A211V mutation in cell cultures and in the CNS of a mutant mouse. The mutation substantially decreased VGLUT3 expression (-70%). We measured VGLUT3-p.A211V activity by vesicular uptake in BON cells, electrophysiological recording of isolated neurons, and its ability to stimulate serotonergic accumulation in cortical synaptic vesicles. Despite a marked loss of expression, the activity of the mutated isoform was only minimally altered. Furthermore, mutant mice displayed none of the behavioral alterations that have previously been reported in VGLUT3 knock-out mice. Finally, we used stimulated emission depletion microscopy to analyze how the mutation altered VGLUT3 distribution within the terminals of mice expressing the mutated isoform. The mutation appeared to reduce the expression of the VGLUT3 transporter by simultaneously decreasing the number of VGLUT3-positive synaptic vesicles and the amount of VGLUT3 per synapses. These observations suggested that VGLUT3 global activity is not linearly correlated with VGLUT3 expression. Furthermore, our data unraveled a nonuniform distribution of VGLUT3 in synaptic vesicles. Identifying the mechanisms responsible for this complex vesicular sorting will be critical to understand VGLUT's involvement in normal and pathological conditions.SIGNIFICANCE STATEMENT VGLUT3 is an atypical member of the vesicular glutamate transporter family. A point mutation of VGLUT3 (VGLUT3-p.A211V) responsible for a progressive loss of hearing has been identified in humans. We observed that this mutation dramatically reduces VGLUT3 expression in terminals (∼70%) without altering its function. Furthermore, using stimulated emission depletion microscopy, we found that reducing the expression levels of VGLUT3 diminished the number of VGLUT3-positive vesicles at synapses. These unexpected findings challenge the vision of a uniform distribution of synaptic vesicles at synapses. Therefore, the overall activity of VGLUT3 is not proportional to the level of VGLUT3 expression. These data will be key in interpreting the role of VGLUTs in human pathologies.


Subject(s)
Brain/metabolism , Point Mutation/physiology , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Proteins/genetics , Vesicular Glutamate Transport Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation
16.
PLoS One ; 11(2): e0148328, 2016.
Article in English | MEDLINE | ID: mdl-26849367

ABSTRACT

We studied the role of testosterone, mediated by the androgen receptor (AR), in modulating temporal order memory for visual objects. For this purpose, we used male mice lacking AR specifically in the nervous system. Control and mutant males were gonadectomized at adulthood and supplemented with equivalent amounts of testosterone in order to normalize their hormonal levels. We found that neural AR deletion selectively impaired the processing of temporal information for visual objects, without affecting classical object recognition or anxiety-like behavior and circulating corticosterone levels, which remained similar to those in control males. Thus, mutant males were unable to discriminate between the most recently seen object and previously seen objects, whereas their control littermates showed more interest in exploring previously seen objects. Because the hippocampal CA1 area has been associated with temporal memory for visual objects, we investigated whether neural AR deletion altered the functionality of this region. Electrophysiological analysis showed that neural AR deletion affected basal glutamate synaptic transmission and decreased the magnitude of N-methyl-D-aspartate receptor (NMDAR) activation and high-frequency stimulation-induced long-term potentiation. The impairment of NMDAR function was not due to changes in protein levels of receptor. These results provide the first evidence for the modulation of temporal processing of information for visual objects by androgens, via AR activation, possibly through regulation of NMDAR signaling in the CA1 area in male mice.


Subject(s)
Hippocampus/physiology , Receptors, Androgen/metabolism , Spatial Processing/physiology , Animals , Anxiety/genetics , Behavior, Animal , Corticosterone/blood , Electrophysiological Phenomena , Gene Deletion , Long-Term Potentiation/physiology , Male , Memory, Short-Term , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity , Receptors, Androgen/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission
17.
AIDS Care ; 28(4): 432-5, 2016.
Article in English | MEDLINE | ID: mdl-26477684

ABSTRACT

Persons living with HIV/AIDS (PLHA) experience clinically significant pain as a result of HIV and such pain is often related to increased levels of anxiety/depression. Pain-related anxiety has been identified as a mechanism in the onset and progression of pain experience and associated affective distress. However, there has not been empirical study of pain-related anxiety in relation to affective processes among PLHA. To address this gap, hierarchical multiple regressions were conducted using SPSS v.21 to examine pain-related anxiety (as measured using the Pain Anxiety Symptoms Scale) in relation to anxiety and depressive symptoms (as measured using the Mood and Anxiety Symptoms Questionnaire) among 93 PLHA (10.8% female; Mean age = 49.63, SD = 8.89). Pain-related anxiety was significantly related to anxious arousal symptoms (ß = .43) and anhedonic depressive symptoms (ß = .25); effects were evident beyond the variance accounted for by CD4 count, race, sex, income level, and current level of bodily pain. The present results suggest that pain-related anxiety may play a role in the experience of anxiety and depressive symptoms among PLHA.


Subject(s)
Anxiety/psychology , Depression/psychology , HIV Infections/psychology , Pain/psychology , Adult , Affect , Anxiety/complications , Cross-Sectional Studies , Depression/complications , Female , HIV Infections/complications , HIV Infections/physiopathology , Humans , Male , Middle Aged , Pain Measurement , Psychiatric Status Rating Scales , Surveys and Questionnaires
18.
Neurobiol Dis ; 87: 69-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26711621

ABSTRACT

Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons. The gold standard therapy relies on dopamine (DA) replacement by the administration of levodopa (l-DOPA). However, with time l-DOPA treatment induces severe motor side effects characterized by abnormal and involuntary movements, or dyskinesia. Although earlier studies point to a role of striatal cholinergic interneurons, also known as striatal tonically active neurons (TANs), in l-DOPA-induced dyskinesia (LID), the underlying mechanisms remain to be fully characterized. Here, we find that DA depletion is accompanied by increased expression of choline acetyltransferase (ChAT), the vesicular acetylcholine transporter (VAChT) as well as the atypical vesicular glutamate transporter type 3 (VGLUT3). TANs number and soma size are not changed. In dyskinetic mice, the VAChT levels remain high whereas the expression of VGLUT3 decreases. LID is attenuated in VGLUT3-deficient mice but not in mice bearing selective inactivation of VAChT in TANs. Finally, the absence of VGLUT3 is accompanied by a reduction of l-DOPA-induced phosphorylation of ERK1/2, ribosomal subunit (rpS6) and GluA1. Our results reveal that VGLUT3 plays an important role in the development of LID and should be considered as a potential and promising therapeutic target for prevention of LID.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/metabolism , Levodopa/toxicity , Amino Acid Transport Systems, Acidic/genetics , Animals , Cell Count , Cell Size , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Dyskinesia, Drug-Induced/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Neurons/pathology , Oxidopamine , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Phosphorylation/drug effects , Receptors, AMPA/metabolism , Ribosomal Protein S6/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
20.
Learn Mem ; 21(5): 287-97, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741109

ABSTRACT

We investigated the specific role of zinc present in large amounts in the synaptic vesicles of mossy fibers and coreleased with glutamate in the CA3 region. In previous studies, we have shown that blockade of zinc after release has no effect on the consolidation of spatial learning, while zinc is required for the consolidation of contextual fear conditioning. Although both are hippocampo-dependent processes, fear conditioning to the context implies a strong emotional burden. To verify the hypothesis that zinc could play a specific role in enabling sustainable memorization of a single event with a strong emotional component, we used a neuropharmacological approach combining a glutamate receptor antagonist with different zinc chelators. Results show that zinc is mandatory to allow the consolidation of one-shot memory, thus being the key element allowing the hippocampus submitted to a strong emotional charge to switch from the cognitive mode to a flashbulb memory mode. Individual differences in learning abilities have been known for a long time to be totally or partially compensated by distributed learning practice. Here we show that contextual fear conditioning impairments due to zinc blockade can be efficiently reduced by distributed learning practice.


Subject(s)
Emotions/physiology , Hippocampus/physiology , Memory/physiology , Mossy Fibers, Hippocampal/metabolism , Zinc/metabolism , Analysis of Variance , Animals , Chelating Agents/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Ditiocarb/pharmacology , Edetic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Freezing Reaction, Cataleptic/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/drug effects , Time Factors , Valine/analogs & derivatives , Valine/pharmacology , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...