Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.529
Filter
1.
Neuron ; 112(3): 488-499.e5, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38086374

ABSTRACT

Ventral tegmental area (VTA) projections to the nucleus accumbens (NAc) drive reward-related motivation. Although dopamine neurons are predominant, a substantial glutamatergic projection is also present, and a subset of these co-release both dopamine and glutamate. Optogenetic stimulation of VTA glutamate neurons not only supports self-stimulation but can also induce avoidance behavior, even in the same assay. Here, we parsed the selective contribution of glutamate or dopamine co-release from VTA glutamate neurons to reinforcement and avoidance. We expressed channelrhodopsin-2 (ChR2) in mouse VTA glutamate neurons in combination with CRISPR-Cas9 to disrupt either the gene encoding vesicular glutamate transporter 2 (VGLUT2) or tyrosine hydroxylase (Th). Selective disruption of VGLUT2 abolished optogenetic self-stimulation but left real-time place avoidance intact, whereas CRISPR-Cas9 deletion of Th preserved self-stimulation but abolished place avoidance. Our results demonstrate that glutamate release from VTA glutamate neurons is positively reinforcing but that dopamine release from VTA glutamate neurons can induce avoidance behavior.


Subject(s)
Dopamine , Glutamic Acid , Mice , Animals , Glutamic Acid/physiology , Reward , Ventral Tegmental Area/physiology , Dopaminergic Neurons/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism , Tyrosine 3-Monooxygenase/metabolism
2.
J Neurosci ; 44(2)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-37989594

ABSTRACT

Glutamate spillover from the synapse is tightly regulated by astrocytes, limiting the activation of extrasynaptically located NMDA receptors (NMDAR). The processes of astrocytes are dynamic and can modulate synaptic physiology. Though norepinephrine (NE) and ß-adrenergic receptor (ß-AR) activity can modify astrocyte volume, this has yet to be confirmed outside of sensory cortical areas, nor has the effect of noradrenergic signaling on glutamate spillover and neuronal NMDAR activity been explored. We monitored changes to astrocyte process volume in response to noradrenergic agonists in the medial prefrontal cortex of male and female mice. Both NE and the ß-AR agonist isoproterenol (ISO) increased process volume by ∼20%, significantly higher than changes seen when astrocytes had G-protein signaling blocked by GDPßS. We measured the effect of ß-AR signaling on evoked NMDAR currents. While ISO did not affect single stimulus excitatory currents of Layer 5 pyramidal neurons, ISO reduced NMDAR currents evoked by 10 stimuli at 50 Hz, which elicits glutamate spillover, by 18%. After isolating extrasynaptic NMDARs by blocking synaptic NMDARs with the activity-dependent NMDAR blocker MK-801, ISO similarly reduced extrasynaptic NMDAR currents in response to 10 stimuli by 18%. Finally, blocking ß-AR signaling in the astrocyte network by loading them with GDPßS reversed the ISO effect on 10 stimuli-evoked NMDAR currents. These results demonstrate that astrocyte ß-AR activity reduces extrasynaptic NMDAR recruitment, suggesting that glutamate spillover is reduced.


Subject(s)
Astrocytes , Receptors, N-Methyl-D-Aspartate , Mice , Animals , Male , Female , Receptors, N-Methyl-D-Aspartate/metabolism , Astrocytes/metabolism , Pyramidal Cells/physiology , Prefrontal Cortex/physiology , Glutamic Acid/physiology , Receptors, Adrenergic, beta , Synapses/physiology
3.
Front Neuroendocrinol ; 70: 101069, 2023 07.
Article in English | MEDLINE | ID: mdl-37149229

ABSTRACT

Hypothalamic melanin-concentrating hormone (MCH) neurons participate in many fundamental neuroendocrine processes. While some of their effects can be attributed to MCH itself, others appear to depend on co-released neurotransmitters. Historically, the subject of fast neurotransmitter co-release from MCH neurons has been contentious, with data to support MCH neurons releasing GABA, glutamate, both, and neither. Rather than assuming a position in that debate, this review considers the evidence for all sides and presents an alternative explanation: neurochemical identity, including classical neurotransmitter content, is subject to change. With an emphasis on the variability of experimental details, we posit that MCH neurons may release GABA and/or glutamate at different points according to environmental and contextual factors. Through the lens of the MCH system, we offer evidence that the field of neuroendocrinology would benefit from a more nuanced and dynamic interpretation of neurotransmitter identity.


Subject(s)
Hypothalamic Hormones , Hypothalamic Hormones/metabolism , Hypothalamic Hormones/pharmacology , Pituitary Hormones/pharmacology , Pituitary Hormones/physiology , Neurons/metabolism , Melanins/pharmacology , Melanins/physiology , Hypothalamus/metabolism , Glutamic Acid/pharmacology , Glutamic Acid/physiology , Neurotransmitter Agents , gamma-Aminobutyric Acid
4.
J Neuroendocrinol ; 35(4): e13253, 2023 04.
Article in English | MEDLINE | ID: mdl-36949648

ABSTRACT

Compared to male pups, perinatal female rats rely heavily on neuronal glutamine (Gln) transport for sustaining glutamatergic synaptic release in neurons of the ventrolateral ventral media nucleus of the hypothalamus (vlVMH). VMH mainly regulates female sexual behavior and increases glutamate release of perinatal hypothalamic neurons, permanently enhances dendrite spine numbers and is associated with brain and behavioral defeminization. We hypothesized that perinatal interruption of neuronal Gln transport may alter the glutamatergic synaptic transmission during adulthood. Perinatal rats of both sexes received an intracerebroventricular injection of a neuronal Gln uptake blocker, alpha-(methylamino) isobutyric acid (MeAIB, 5 mM), and were raised until adulthood. Whole-cell voltage-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and evoked EPSCs (eEPSCs) of vlVMH neurons in adult rats with the perinatal pretreatment were conducted and neuron morphology was subjected to post hoc examination. Perinatal MeAIB treatment sex-differentially increased mEPSC frequency in males, but decreased mEPSC amplitude and synaptic Glu release in females. The pretreatment sex-differentially decreased eEPSC amplitude in males but increased AMPA/NMDA current ratio in females, and changed the morphology of vlVMH neurons of adult rats to that of the opposite sex. Most alterations in the glutamatergic synaptic transmission resembled the changes occurring during MeAIB acute exposure in perinatal rats of both sexes. We conclude that perinatal blockade of neuronal Gln transport mediates changes via different presynaptic and postsynaptic mechanisms to induce sex-differential alterations of the glutamatergic synaptic transmission and organization of vlVMH neurons in adult rats. These changes may be permanent and associated with brain and behavior feminization and/or defeminization in rats.


Subject(s)
Glutamine , Neurons , Pregnancy , Rats , Animals , Male , Female , Rats, Sprague-Dawley , Synaptic Transmission/physiology , Glutamic Acid/physiology , Hypothalamus
5.
Prog Mol Biol Transl Sci ; 196: 59-97, 2023.
Article in English | MEDLINE | ID: mdl-36813366

ABSTRACT

AMPA receptors are glutamate-gated ion channels, present in a wide range of neuron types and in glial cells. Their main role is to mediate fast excitatory synaptic transmission, and therefore, they are critical for normal brain function. In neurons, AMPA receptors undergo constitutive and activity-dependent trafficking between the synaptic, extrasynaptic and intracellular pools. The kinetics of AMPA receptor trafficking is crucial for the precise functioning of both individual neurons and neural networks involved in information processing and learning. Many of the neurological diseases evoked by neurodevelopmental and neurodegenerative malfunctions or traumatic injuries are caused by impaired synaptic function in the central nervous system. For example, attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD), tumors, seizures, ischemic strokes, and traumatic brain injury are all characterized by impaired glutamate homeostasis and associated neuronal death, typically caused by excitotoxicity. Given the important role of AMPA receptors in neuronal function, it is not surprising that perturbations in AMPA receptor trafficking are associated with these neurological disorders. In this book chapter, we will first introduce the structure, physiology and synthesis of AMPA receptors, followed by an in-depth description of the molecular mechanisms that control AMPA receptor endocytosis and surface levels under basal conditions or synaptic plasticity. Finally, we will discuss how impairments in AMPA receptor trafficking, particularly endocytosis, contribute to the pathophysiology of various neurological disorders and what efforts are being made to therapeutically target this process.


Subject(s)
Nervous System Diseases , Receptors, AMPA , Humans , Receptors, AMPA/metabolism , Synaptic Transmission , Glutamic Acid/physiology , Endocytosis
6.
J Neurosci ; 42(46): 8670-8693, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36195440

ABSTRACT

We identified three types of monosynaptic cholinergic inputs spatially arranged onto medial substantia nigra dopaminergic neurons in male and female mice: cotransmitted acetylcholine (ACh)/GABA, GABA-only, and ACh only. There was a predominant GABA-only conductance along lateral dendrites and soma-centered ACh/GABA cotransmission. In response to repeated stimulation, the GABA conductance found on lateral dendrites decremented less than the proximally located GABA conductance, and was more effective at inhibiting action potentials. While soma-localized ACh/GABA cotransmission showed depression of the GABA component with repeated stimulation, ACh-mediated nicotinic responses were largely maintained. We investigated whether this differential change in inhibitory/excitatory inputs leads to altered neuronal excitability. We found that a depolarizing current or glutamate preceded by cotransmitted ACh/GABA was more effective in eliciting an action potential compared with current, glutamate, or ACh/GABA alone. This enhanced excitability was abolished with nicotinic receptor inhibitors, and modulated by T- and L-type calcium channels, thus establishing that activity of multiple classes of ion channels integrates to shape neuronal excitability.SIGNIFICANCE STATEMENT Our laboratory has previously discovered a population of substantia nigra dopaminegic neurons (DA) that receive cotransmitted ACh and GABA. This study used subcellular optogenetic stimulation of cholinergic presynaptic terminals to map the functional ACh and GABA synaptic inputs across the somatodendritic extent of substantia nigra DA neurons. We determined spatially clustered GABA-only inputs on the lateral dendrites while cotransmitted ACh and GABA clustered close to the soma. We have shown that the action of GABA and ACh in cotransmission spatially clustered near the soma play a critical role in enhancing glutamate-mediated neuronal excitability through the activation of T- and L-type voltage-gated calcium channels.


Subject(s)
Acetylcholine , Dopaminergic Neurons , Male , Female , Mice , Animals , Acetylcholine/pharmacology , Glutamic Acid/physiology , Cholinergic Agents , gamma-Aminobutyric Acid , Synaptic Transmission/physiology
7.
Neuron ; 110(12): 1993-2008.e6, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35443154

ABSTRACT

Empathic pain has attracted the interest of a substantial number of researchers studying the social transfer of pain in the sociological, psychological, and neuroscience fields. However, the neural mechanism of empathic pain remains elusive. Here, we establish a long-term observational pain model in mice and find that glutamatergic projection from the insular cortex (IC) to the basolateral amygdala (BLA) is critical for the formation of observational pain. The selective activation or inhibition of the IC-BLA projection pathway strengthens or weakens the intensity of observational pain, respectively. The synaptic molecules are screened, and the upregulated synaptotagmin-2 and RIM3 are identified as key signals in controlling the increased synaptic glutamate transmission from the IC to the BLA. Together, these results reveal the molecular and synaptic mechanisms of a previously unidentified neural pathway that regulates observational pain in mice.


Subject(s)
Basolateral Nuclear Complex , Animals , Basolateral Nuclear Complex/physiology , Cerebral Cortex/physiology , Glutamic Acid/physiology , Insular Cortex , Mice , Pain , Synapses
8.
Mol Brain ; 14(1): 152, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34607601

ABSTRACT

The glutamatergic signaling pathway is involved in molecular learning and human cognitive ability. Specific single variants (SNVs, formerly single-nucleotide polymorphisms) in the genes encoding N-methyl-D-aspartate receptor subunits have been associated with neuropsychiatric disorders by altering glutamate transmission. However, these variants associated with cognition and mental activity have rarely been explored in healthy adolescents. In this study, we screened for SNVs in the glutamatergic signaling pathway to identify genetic variants associated with cognitive ability. We found that SNVs in the subunits of ionotropic glutamate receptors, including GRIA1, GRIN1, GRIN2B, GRIN2C, GRIN3A, GRIN3B, and calcium/calmodulin-dependent protein kinase IIα (CaMK2A) are associated with cognitive function. Plasma CaMK2A level was correlated positively with the cognitive ability of Taiwanese senior high school students. We demonstrated that elevating CaMK2A increased its autophosphorylation at T286 and increased the expression of its downstream targets, including GluA1 and phosphor- GluA1 in vivo. Additionally, methyl-CpG binding protein 2 (MeCP2), a downstream target of CaMK2A, was found to activate the expression of CaMK2A, suggesting that MeCP2 and CaMK2A can form a positive feedback loop. In summary, two members of the glutamatergic signaling pathway, CaMK2A and MeCP2, are implicated in the cognitive ability of adolescents; thus, altering the expression of CaMK2A may affect cognitive ability in youth.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cognition/physiology , Methyl-CpG-Binding Protein 2/physiology , Psychology, Adolescent , Receptors, Ionotropic Glutamate/genetics , Signal Transduction/physiology , Adolescent , Calcium-Calmodulin-Dependent Protein Kinase Type 2/blood , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line, Tumor , Enzyme Activation , Feedback, Physiological/physiology , Female , Glutamic Acid/physiology , HEK293 Cells , Humans , Male , Neuroblastoma , Phosphorylation , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Processing, Post-Translational , Receptors, Ionotropic Glutamate/physiology , Reference Values , Taiwan
9.
Mol Brain ; 14(1): 143, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34530877

ABSTRACT

Vacuolar protein sorting 35 (VPS35) regulates neurotransmitter receptor recycling from endosomes. A missense mutation (D620N) in VPS35 leads to autosomal-dominant, late-onset Parkinson's disease. Here, we study the basic neurobiology of VPS35 and Parkinson's disease mutation effects in the D620N knock-in mouse and the effect of leucine-rich repeat kinase 2 (LRRK2) inhibition on synaptic phenotypes. The study was conducted using a VPS35 D620N knock-in mouse that expresses VPS35 at endogenous levels. Protein levels, phosphorylation states, and binding ratios in brain lysates from knock-in mice and wild-type littermates were assayed by co-immunoprecipitation and western blot. Dendritic protein co-localization, AMPA receptor surface expression, synapse density, and glutamatergic synapse activity in primary cortical cultures from knock-in and wild-type littermates were assayed using immunocytochemistry and whole-cell patch clamp electrophysiology. In brain tissue, we confirm VPS35 forms complexes with LRRK2 and AMPA-type glutamate receptor GluA1 subunits, in addition to NMDA-type glutamate receptor GluN1 subunits and D2-type dopamine receptors. Receptor and LRRK2 binding was unaltered in D620N knock-in mice, but we confirm the mutation results in reduced binding of VPS35 with WASH complex member FAM21, and increases phosphorylation of the LRRK2 kinase substrate Rab10, which is reversed by LRRK2 kinase inhibition in vivo. In cultured cortical neurons from knock-in mice, pRab10 is also increased, and reversed by LRRK2 inhibition. The mutation also results in increased endosomal recycling protein cluster density (VPS35-FAM21 co-clusters and Rab11 clusters), glutamate transmission, and GluA1 surface expression. LRRK2 kinase inhibition, which reversed Rab10 hyper-phosphorylation, did not rescue elevated glutamate release or surface GluA1 expression in knock-in neurons, but did alter AMPAR traffic in wild-type cells. The results improve our understanding of the cell biology of VPS35, and the consequences of the D620N mutation in developing neuronal networks. Together the data support a chronic synaptopathy model for latent neurodegeneration, providing phenotypes and candidate pathophysiological stresses that may drive eventual transition to late-stage parkinsonism in VPS35 PD. The study demonstrates the VPS35 mutation has effects that are independent of ongoing LRRK2 kinase activity, and that LRRK2 kinase inhibition alters basal physiology of glutamate synapses in vitro.


Subject(s)
Endosomes/physiology , Glutamic Acid/physiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Mutation, Missense , Parkinson Disease/genetics , Point Mutation , Vesicular Transport Proteins/genetics , Animals , Cells, Cultured , Dendrites/metabolism , Gain of Function Mutation , Gene Knock-In Techniques , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/physiology , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/physiology , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Protein Binding , Protein Interaction Mapping , Receptors, AMPA/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synapses/metabolism , Vesicular Transport Proteins/physiology , rab GTP-Binding Proteins/metabolism
10.
Neuropharmacology ; 198: 108725, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34375625

ABSTRACT

Reinforcement, reward, and aversion are fundamental processes for guiding appropriate behaviors. Longstanding theories have pointed to dopaminergic neurons of the ventral tegmental area (VTA) and the limbic systems' descending pathways as crucial systems for modulating these behaviors. The application of optogenetic techniques in neurotransmitter- and projection-specific circuits has supported and enhanced many preexisting theories but has also revealed many unexpected results. Here, we review the past decade of optogenetic experiments to study the neural circuitry of reinforcement and reward/aversion with a focus on the mesolimbic dopamine system and brain areas along the medial forebrain bundle (MFB). The cumulation of these studies to date has revealed generalizable findings across molecularly defined cell types in areas of the basal forebrain and anterior hypothalamus. Optogenetic stimulation of GABAergic neurons in these brain regions drives reward and can support positive reinforcement and optogenetic stimulation of glutamatergic neurons in these regions drives aversion. We also review studies of the activity dynamics of neurotransmitter defined populations in these areas which have revealed varied response patterns associated with motivated behaviors. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Subject(s)
Glutamic Acid/physiology , Nerve Net/physiology , Reward , gamma-Aminobutyric Acid/physiology , Animals , Avoidance Learning , GABAergic Neurons , Humans
11.
Neuropharmacology ; 199: 108740, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34343611

ABSTRACT

Glutamate (Glu) is the primary excitatory transmitter in the mammalian brain. But, we know little about the evolutionary history of this adaptation, including the selection of l-glutamate as a signaling molecule in the first place. Here, we used comparative metabolomics and genomic data to reconstruct the genealogy of glutamatergic signaling. The origin of Glu-mediated communications might be traced to primordial nitrogen and carbon metabolic pathways. The versatile chemistry of L-Glu placed this molecule at the crossroad of cellular biochemistry as one of the most abundant metabolites. From there, innovations multiplied. Many stress factors or injuries could increase extracellular glutamate concentration, which led to the development of modular molecular systems for its rapid sensing in bacteria and archaea. More than 20 evolutionarily distinct families of ionotropic glutamate receptors (iGluRs) have been identified in eukaryotes. The domain compositions of iGluRs correlate with the origins of multicellularity in eukaryotes. Although L-Glu was recruited as a neuro-muscular transmitter in the early-branching metazoans, it was predominantly a non-neuronal messenger, with a possibility that glutamatergic synapses evolved more than once. Furthermore, the molecular secretory complexity of glutamatergic synapses in invertebrates (e.g., Aplysia) can exceed their vertebrate counterparts. Comparative genomics also revealed 15+ subfamilies of iGluRs across Metazoa. However, most of this ancestral diversity had been lost in the vertebrate lineage, preserving AMPA, Kainate, Delta, and NMDA receptors. The widespread expansion of glutamate synapses in the cortical areas might be associated with the enhanced metabolic demands of the complex brain and compartmentalization of Glu signaling within modular neuronal ensembles.


Subject(s)
Biological Evolution , Glutamic Acid/physiology , Receptors, Glutamate/physiology , Signal Transduction/physiology , Synapses/physiology , Animals
12.
Anesthesiology ; 135(4): 633-648, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34270686

ABSTRACT

BACKGROUND: Parabrachial nucleus excitation reduces cortical delta oscillation (0.5 to 4 Hz) power and recovery time associated with anesthetics that enhance γ-aminobutyric acid type A receptor action. The effects of parabrachial nucleus excitation on anesthetics with other molecular targets, such as dexmedetomidine and ketamine, remain unknown. The hypothesis was that parabrachial nucleus excitation would cause arousal during dexmedetomidine and ketamine anesthesia. METHODS: Designer Receptors Exclusively Activated by Designer Drugs were used to excite calcium/calmodulin-dependent protein kinase 2α-positive neurons in the parabrachial nucleus region of adult male rats without anesthesia (nine rats), with dexmedetomidine (low dose: 0.3 µg · kg-1 · min-1 for 45 min, eight rats; high dose: 4.5 µg · kg-1 · min-1 for 10 min, seven rats), or with ketamine (low dose: 2 mg · kg-1 · min-1 for 30 min, seven rats; high dose: 4 mg · kg-1 · min-1 for 15 min, eight rats). For control experiments (same rats and treatments), the Designer Receptors Exclusively Activated by Designer Drugs were not excited. The electroencephalogram and anesthesia recovery times were recorded and analyzed. RESULTS: Parabrachial nucleus excitation reduced delta power in the prefrontal electroencephalogram with low-dose dexmedetomidine for the 150-min analyzed period, excepting two brief periods (peak median bootstrapped difference [clozapine-N-oxide - saline] during dexmedetomidine infusion = -6.06 [99% CI = -12.36 to -1.48] dB, P = 0.007). However, parabrachial nucleus excitation was less effective at reducing delta power with high-dose dexmedetomidine and low- and high-dose ketamine (peak median bootstrapped differences during high-dose [dexmedetomidine, ketamine] infusions = [-1.93, -0.87] dB, 99% CI = [-4.16 to -0.56, -1.62 to -0.18] dB, P = [0.006, 0.019]; low-dose ketamine had no statistically significant decreases during the infusion). Recovery time differences with parabrachial nucleus excitation were not statistically significant for dexmedetomidine (median difference for [low, high] dose = [1.63, 11.01] min, 95% CI = [-20.06 to 14.14, -20.84 to 23.67] min, P = [0.945, 0.297]) nor low-dose ketamine (median difference = 12.82 [95% CI: -3.20 to 39.58] min, P = 0.109) but were significantly longer for high-dose ketamine (median difference = 11.38 [95% CI: 1.81 to 24.67] min, P = 0.016). CONCLUSIONS: These results suggest that the effectiveness of parabrachial nucleus excitation to change the neurophysiologic and behavioral effects of anesthesia depends on the anesthetic's molecular target.


Subject(s)
Delta Rhythm/drug effects , Dexmedetomidine/pharmacology , Glutamic Acid , Ketamine/pharmacology , Neurons/drug effects , Parabrachial Nucleus/drug effects , Anesthesia/methods , Anesthetics, Dissociative/pharmacology , Animals , Calcium-Binding Proteins/physiology , Delta Rhythm/physiology , Glutamic Acid/physiology , Hypnotics and Sedatives/pharmacology , Male , Neurons/physiology , Parabrachial Nucleus/physiology , Rats , Rats, Sprague-Dawley
13.
Neural Plast ; 2021: 4784385, 2021.
Article in English | MEDLINE | ID: mdl-34306060

ABSTRACT

Noise overexposure leads to hair cell loss, synaptic ribbon reduction, and auditory nerve deterioration, resulting in transient or permanent hearing loss depending on the exposure severity. Oxidative stress, inflammation, calcium overload, glutamate excitotoxicity, and energy metabolism disturbance are the main contributors to noise-induced hearing loss (NIHL) up to now. Gene variations are also identified as NIHL related. Glucocorticoid is the only approved medication for NIHL treatment. New pharmaceuticals targeting oxidative stress, inflammation, or noise-induced neuropathy are emerging, highlighted by the nanoparticle-based drug delivery system. Given the complexity of the pathogenesis behind NIHL, deeper and more comprehensive studies still need to be fulfilled.


Subject(s)
Hearing Loss, Noise-Induced/etiology , Animals , Autophagy , Calcium/metabolism , Clinical Trials, Phase II as Topic , DNA Repair/genetics , Drugs, Investigational/therapeutic use , Energy Metabolism , Gap Junctions , Glutamic Acid/physiology , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/epidemiology , Hearing Loss, Noise-Induced/genetics , Humans , Inflammation , Isoindoles/therapeutic use , Nanoparticles , Organoselenium Compounds/therapeutic use , Oxidative Stress , Potassium Channels/genetics , Stereocilia/ultrastructure
14.
Neuropharmacology ; 197: 108727, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34314736

ABSTRACT

As for electronic computation, neural information processing is energetically expensive. This is because information is coded in the brain as membrane voltage changes, which are generated largely by passive ion movements down electrochemical gradients, and these ion movements later need to be reversed by active ATP-dependent ion pumping. This article will review how much of the energetic cost of the brain reflects the activity of glutamatergic synapses, consider the relative amount of energy used pre- and postsynaptically, outline how evolution has energetically optimised synapse function by adjusting the presynaptic release probability and the postsynaptic number of glutamate receptors, and speculate on how energy use by synapses may be sensed and adjusted. This article is part of the special Issue on 'Glutamate Receptors - The Glutamatergic Synapse'.


Subject(s)
Energy Metabolism/physiology , Glutamic Acid/physiology , Synapses/metabolism , Adenosine Triphosphate/metabolism , Animals , Electrophysiological Phenomena , Energy Metabolism/drug effects , Humans
15.
Neurobiol Dis ; 158: 105448, 2021 10.
Article in English | MEDLINE | ID: mdl-34280523

ABSTRACT

INTRODUCTION: Plasticity at corticostriatal synapses is a key substrate for a variety of brain functions - including motor control, learning and reward processing - and is often disrupted in disease conditions. Despite intense research pointing toward a dynamic interplay between glutamate, dopamine (DA), and serotonin (5-HT) neurotransmission, their precise circuit and synaptic mechanisms regulating their role in striatal plasticity are still unclear. Here, we analyze the role of serotonergic raphe-striatal innervation in the regulation of DA-dependent corticostriatal plasticity. METHODS: Mice (males and females, 2-6 months of age) were housed in standard plexiglass cages at constant temperature (22 ± 1°C) and maintained on a 12/12h light/dark cycle with food and demineralized water ad libitum. In the present study, we used a knock-in mouse line in which the green fluorescent protein reporter gene (GFP) replaced the I Tph2 exon (Tph2GFP mice), allowing selective expression of GFP in the whole 5-HT system, highlighting both somata and neuritis of serotonergic neurons. Heterozygous, Tph2+/GFP, mice were intercrossed to obtain experimental cohorts, which included Wild-type (Tph2+/+), Heterozygous (Tph2+/GFP), and Mutant serotonin-depleted (Tph2GFP/GFP) animals. RESULTS: Using male and female mice, carrying on different Tph2 gene dosages, we show that Tph2 gene modulation results in sex-specific corticostriatal abnormalities, encompassing the abnormal amplitude of spontaneous glutamatergic transmission and the loss of Long Term Potentiation (LTP) in Tph2GFP/GFP mice of both sexes, while this form of plasticity is normally expressed in control mice (Tph2+/+). Once LTP is induced, only the Tph2+/GFP female mice present a loss of synaptic depotentiation. CONCLUSION: We showed a relevant role of the interaction between dopaminergic and serotonergic systems in controlling striatal synaptic plasticity. Overall, our data unveil that 5-HT plays a primary role in regulating DA-dependent corticostriatal plasticity in a sex-related manner and propose altered 5-HT levels as a critical determinant of disease-associated plasticity defects.


Subject(s)
Neostriatum/physiology , Neuronal Plasticity/physiology , Serotonin/physiology , Synapses/physiology , Animals , Animals, Genetically Modified , Electrophysiological Phenomena , Female , Glutamic Acid/physiology , Long-Term Potentiation , Male , Mice , Nerve Fibers , Parkinson Disease, Secondary/physiopathology , Sex Characteristics , Synaptic Transmission/physiology , Tryptophan Hydroxylase/metabolism
16.
Nat Commun ; 12(1): 2811, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990558

ABSTRACT

The supramammillary region (SuM) is a posterior hypothalamic structure, known to regulate hippocampal theta oscillations and arousal. However, recent studies reported that the stimulation of SuM neurons with neuroactive chemicals, including substances of abuse, is reinforcing. We conducted experiments to elucidate how SuM neurons mediate such effects. Using optogenetics, we found that the excitation of SuM glutamatergic (GLU) neurons was reinforcing in mice; this effect was relayed by their projections to septal GLU neurons. SuM neurons were active during exploration and approach behavior and diminished activity during sucrose consumption. Consistently, inhibition of SuM neurons disrupted approach responses, but not sucrose consumption. Such functions are similar to those of mesolimbic dopamine neurons. Indeed, the stimulation of SuM-to-septum GLU neurons and septum-to-ventral tegmental area (VTA) GLU neurons activated mesolimbic dopamine neurons. We propose that the supramammillo-septo-VTA pathway regulates arousal that reinforces and energizes behavioral interaction with the environment.


Subject(s)
Dopaminergic Neurons/physiology , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Consummatory Behavior/drug effects , Consummatory Behavior/physiology , Dopamine/physiology , Female , Glutamic Acid/physiology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Rats , Rats, Wistar , Reinforcement, Psychology , Septum of Brain/cytology , Septum of Brain/drug effects , Septum of Brain/physiology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage
17.
J Neurochem ; 158(2): 182-196, 2021 07.
Article in English | MEDLINE | ID: mdl-33894004

ABSTRACT

Calcium-dependent activator protein for secretion 1 (CAPS1) is a SNARE accessory protein that facilitates formation of the SNARE complex to enable neurotransmitter release. Messenger RNAs encoding CAPS1 are subject to a site-specific adenosine-to-inosine (A-to-I) editing event resulting in a glutamate-to-glycine (E-to-G) substitution in the C-terminal domain of the encoded protein product. The C-terminal domain of CAPS1 is necessary for its synaptic enrichment and Cadps RNA editing has been shown previously to enhance the release of neuromodulatory transmitters. Using mutant mouse lines engineered to solely express CAPS1 protein isoforms encoded by either the non-edited or edited Cadps transcript, primary neuronal cultures from mouse hippocampus were used to explore the effect of Cadps editing on neurotransmission and CAPS1 synaptic localization at both glutamatergic and GABAergic synapses. While the editing of Cadps does not alter baseline evoked neurotransmission, it enhances short-term synaptic plasticity, specifically short-term depression, at inhibitory synapses. Cadps editing also alters spontaneous inhibitory neurotransmission. Neurons that solely express edited Cadps have a greater proportion of synapses that contain CAPS1 than neurons that solely express non-edited Cadps for both glutamatergic and GABAergic synapses. Editing of Cadps transcripts is regulated by neuronal activity, as global network stimulation increases the extent of transcripts edited in wild-type hippocampal neurons, whereas chronic network silencing decreases the level of Cadps editing. Taken together, these results provide key insights into the importance of Cadps editing in modulating its own synaptic localization, as well as the modulation of neurotransmission at inhibitory synapses in hippocampal neurons.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , RNA Editing/genetics , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Adenosine Deaminase/metabolism , Animals , CRISPR-Cas Systems , Electrophysiological Phenomena , Glutamic Acid/physiology , High-Throughput Nucleotide Sequencing , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mutation , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Primary Cell Culture , RNA Editing/physiology , gamma-Aminobutyric Acid/physiology
18.
Physiol Rep ; 9(6): e14774, 2021 03.
Article in English | MEDLINE | ID: mdl-33769694

ABSTRACT

N-methyl-d-aspartate (NMDA) application has conventionally been used to activate spinal networks to induce locomotion in spinalized animals. We recently described an alternative approach in which application of continuous blue light activates channelrhodopsin-2 in vesicular glutamate transporter 2a (vglut2a)-expressing spinal neurons to produce organized, rhythmic locomotor activity in spinally-transected larval zebrafish. This technique arguably enhances research validity, because endogenous glutamate is released into existing synapses instead of activating only a subset of glutamatergic (NMDA) receptors with an exogenous compound. Here, we explored the viability of this approach in the context of using it for longer-term experiments. Fictive swimming was induced through repetitive application of 10-s blue light stimuli to spinalized preparations for up to 60 min at intervals of 1, 3, or 15 min. Locomotor activity was maintained throughout the experimental timecourse, demonstrating the robustness of the system. Although locomotor bursts remained organized into episodes of activity, the number of bursts elicited during each successive stimulus decreased. This was in contrast to NMDA bath application, in which bursts became less episodically organized while the overall number of bursts remained unchanged. The efficacy of the repetitive optogenetic stimulation paradigm was demonstrated through application of exogenous dopamine, which reversibly decreased the number of bursts produced per stimulus compared with untreated preparations. Finally, increasing the stimulus interval to 15 min lessened, but did not eliminate locomotor fatigue from repetitive activation. Altogether, we established repetitive optogenetic stimulation of vglut2a-expressing neurons as a viable alternative to NMDA application for activation of the zebrafish spinal locomotor network.


Subject(s)
Glutamic Acid/physiology , Locomotion/physiology , Motor Neurons/physiology , N-Methylaspartate/physiology , Neurons/physiology , Optogenetics , Spinal Cord/physiology , Animals , Excitatory Amino Acid Agonists/administration & dosage , Fatigue , Locomotion/drug effects , Models, Animal , Motor Neurons/drug effects , N-Methylaspartate/administration & dosage , Neurons/drug effects , Spinal Cord/drug effects , Swimming , Zebrafish
19.
Mol Cell Neurosci ; 112: 103613, 2021 04.
Article in English | MEDLINE | ID: mdl-33753311

ABSTRACT

Presynaptic neurotransmitter release is strictly regulated by SNARE proteins, Ca2+ and a number of Ca2+ sensors including synaptotagmins (Syts) and Double C2 domain proteins (Doc2s). More than seventy years after the original description of spontaneous release, the mechanism that regulates this process is still poorly understood. Syt-1, Syt7 and Doc2 proteins contribute predominantly, but not exclusively, to synchronous, asynchronous and spontaneous phases of release. The proteins share a conserved tandem C2 domain architecture, but are functionally diverse in their subcellular location, Ca2+-binding properties and protein interactions. In absence of Syt-1, Doc2a and -b, neurons still exhibit spontaneous vesicle fusion which remains Ca2+-sensitive, suggesting the existence of additional sensors. Here, we selected Doc2c, rabphilin-3a and Syt-7 as three potential Ca2+ sensors for their sequence homology with Syt-1 and Doc2b. We genetically ablated each candidate gene in absence of Doc2a and -b and investigated spontaneous and evoked release in glutamatergic hippocampal neurons, cultured either in networks or on microglial islands (autapses). The removal of Doc2c had no effect on spontaneous or evoked release. Syt-7 removal also did not affect spontaneous release, although it altered short-term plasticity by accentuating short-term depression. The removal of rabphilin caused an increased spontaneous release frequency in network cultures, an effect that was not observed in autapses. Taken together, we conclude that Doc2c and Syt-7 do not affect spontaneous release of glutamate in hippocampal neurons, while our results suggest a possible regulatory role of rabphilin-3a in neuronal networks. These findings importantly narrow down the repertoire of synaptic Ca2+ sensors that may be implicated in the spontaneous release of glutamate.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Calcium-Binding Proteins/physiology , Calcium/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Synaptotagmin I/physiology , Vesicular Transport Proteins/physiology , Action Potentials , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cells, Cultured , Conserved Sequence , Glutamic Acid/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Protein Domains , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Synaptotagmin I/chemistry , Synaptotagmin I/deficiency , Synaptotagmin I/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/deficiency , Vesicular Transport Proteins/genetics , Rabphilin-3A
20.
Neuron ; 109(7): 1150-1167.e6, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33600763

ABSTRACT

The hypothalamus plays crucial roles in regulating endocrine, autonomic, and behavioral functions via its diverse nuclei and neuronal subtypes. The developmental mechanisms underlying ontogenetic establishment of different hypothalamic nuclei and generation of neuronal diversity remain largely unknown. Here, we show that combinatorial T-box 3 (TBX3), orthopedia homeobox (OTP), and distal-less homeobox (DLX) expression delineates all arcuate nucleus (Arc) neurons and defines four distinct subpopulations, whereas combinatorial NKX2.1/SF1 and OTP/DLX expression identifies ventromedial hypothalamus (VMH) and tuberal nucleus (TuN) neuronal subpopulations, respectively. Developmental analysis indicates that all four Arc subpopulations are mosaically and simultaneously generated from embryonic Arc progenitors, whereas glutamatergic VMH neurons and GABAergic TuN neurons are sequentially generated from common embryonic VMH progenitors. Moreover, clonal lineage-tracing analysis reveals that diverse lineages from multipotent radial glia progenitors orchestrate Arc and VMH-TuN establishment. Together, our study reveals cellular mechanisms underlying generation and organization of diverse neuronal subtypes and ontogenetic establishment of individual nuclei in the mammalian hypothalamus.


Subject(s)
Hypothalamus/cytology , Hypothalamus/growth & development , Neurons/physiology , Animals , Animals, Genetically Modified , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/embryology , Cell Lineage , Glutamic Acid/physiology , Homeodomain Proteins/metabolism , Hypothalamus/embryology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Stem Cells/physiology , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/embryology , Ventromedial Hypothalamic Nucleus/metabolism , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...