Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
1.
J Neurosci ; 41(35): 7329-7339, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34290081

ABSTRACT

Post-tetanic potentiation (PTP) is a form of short-term plasticity that lasts for tens of seconds following a burst of presynaptic activity. It has been proposed that PTP arises from protein kinase C (PKC) phosphorylation of Munc18-1, an SM (Sec1/Munc-18 like) family protein that is essential for release. To test this model, we made a knock-in mouse in which all Munc18-1 PKC phosphorylation sites were eliminated through serine-to-alanine point mutations (Munc18-1SA mice), and we studied mice of either sex. The expression of Munc18-1 was not altered in Munc18-1SA mice, and there were no obvious behavioral phenotypes. At the hippocampal CA3-to-CA1 synapse and the granule cell parallel fiber (PF)-to-Purkinje cell (PC) synapse, basal transmission was largely normal except for small decreases in paired-pulse facilitation that are consistent with a slight elevation in release probability. Phorbol esters that mimic the activation of PKC by diacylglycerol still increased synaptic transmission in Munc18-1SA mice. In Munc18-1SA mice, 70% of PTP remained at CA3-to-CA1 synapses, and the amplitude of PTP was not reduced at PF-to-PC synapses. These findings indicate that at both CA3-to-CA1 and PF-to-PC synapses, phorbol esters and PTP enhance synaptic transmission primarily by mechanisms that are independent of PKC phosphorylation of Munc18-1.SIGNIFICANCE STATEMENT A leading mechanism for a prevalent form of short-term plasticity, post-tetanic potentiation (PTP), involves protein kinase C (PKC) phosphorylation of Munc18-1. This study tests this mechanism by creating a knock-in mouse in which Munc18-1 is replaced by a mutated form of Munc18-1 that cannot be phosphorylated. The main finding is that most PTP at hippocampal CA3-to-CA1 synapses or at cerebellar granule cell-to-Purkinje cell synapses does not rely on PKC phosphorylation of Munc18-1. Thus, mechanisms independent of PKC phosphorylation of Munc18-1 are important mediators of PTP.


Subject(s)
Munc18 Proteins/metabolism , Neuronal Plasticity/physiology , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Female , Gene Knock-In Techniques , Hippocampus/physiology , Male , Mice , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Munc18 Proteins/deficiency , Mutation, Missense , Phorbol Esters/pharmacology , Phosphorylation , Point Mutation , Protein Kinase C/deficiency , Purkinje Cells/physiology , Recombinant Proteins/metabolism , Synaptic Transmission/drug effects
2.
J Neuroinflammation ; 18(1): 279, 2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34857006

ABSTRACT

BACKGROUND: Opioid analgesics remain widely used for pain treatment despite the related serious side effects. Some of those, such as opioid tolerance and opioid-induced hyperalgesia may be at least partially due to modulation of opioid receptors (OR) function at nociceptive synapses in the spinal cord dorsal horn. It was suggested that increased release of different chemokines under pathological conditions may play a role in this process. The goal of this study was to investigate the crosstalk between the µOR, transient receptor potential vanilloid 1 (TRPV1) receptor and C-C motif ligand 2 (CCL2) chemokine and the involvement of spinal microglia in the modulation of opioid analgesia. METHODS: Patch-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and dorsal root evoked currents (eEPSC) in spinal cord slices superficial dorsal horn neurons were used to evaluate the effect of µOR agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), CCL2, TRPV1 antagonist SB366791 and minocycline. Paw withdrawal test to thermal stimuli was combined with intrathecal (i.t.) delivery of CCL2 and DAMGO to investigate the modulation in vivo. RESULTS: Application of DAMGO induced a rapid decrease of mEPSC frequency and eEPSC amplitude, followed by a delayed increase of the eESPC amplitude, which was prevented by SB366791. Chemokine CCL2 treatment significantly diminished all the DAMGO-induced changes. Minocycline treatment prevented the CCL2 effects on the DAMGO-induced eEPSC depression, while mEPSC changes were unaffected. In behavioral experiments, i.t. injection of CCL2 completely blocked DAMGO-induced thermal hypoalgesia and intraperitoneal pre-treatment with minocycline prevented the CCL2 effect. CONCLUSIONS: Our results indicate that opioid-induced inhibition of the excitatory synaptic transmission could be severely attenuated by increased CCL2 levels most likely through a microglia activation-dependent mechanism. Delayed potentiation of neurotransmission after µOR activation is dependent on TRPV1 receptors activation. Targeting CCL2 and its receptors and TRPV1 receptors in combination with opioid therapy could significantly improve the analgesic properties of opioids, especially during pathological states.


Subject(s)
Analgesics, Opioid/pharmacology , Chemokine CCL2/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Nociception/drug effects , Spinal Cord Dorsal Horn/drug effects , Spinal Cord/drug effects , Synaptic Transmission/drug effects , Anilides/pharmacology , Animals , Cinnamates/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Male , Miniature Postsynaptic Potentials/drug effects , Neurons/drug effects , Rats , Rats, Wistar
3.
J Neurosci ; 39(25): 4909-4930, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30971438

ABSTRACT

Although NPY has potent anxiolytic actions within the BLA, selective activation of BLA NPY Y2 receptors (Y2Rs) acutely increases anxiety by an unknown mechanism. Using ex vivo male rat brain slice electrophysiology, we show that the selective Y2R agonist, [ahx5-24]NPY, reduced the frequency of GABAA-mediated mIPSCs in BLA principal neurons (PNs). [ahx5-24]NPY also reduced tonic activation of GABAB receptors (GABABR), which increased PN excitability through inhibition of a tonic, inwardly rectifying potassium current (KIR ). Surprisingly, Y2R-sensitive GABABR currents were action potential-independent, persisting after treatment with TTX. Additionally, the Ca2+-dependent, slow afterhyperpolarizing K+ current (IsAHP ) was enhanced in approximately half of the Y2R-sensitive PNs, possibly from enhanced Ca2+ influx, permitted by reduced GABABR tone. In male and female mice expressing tdTomato in Y2R-mRNA cells (tdT-Y2R mice), immunohistochemistry revealed that BLA somatostatin interneurons express Y2Rs, as do a significant subset of BLA PNs. In tdT-Y2R mice, [ahx5-24]NPY increased excitability and suppressed the KIR in nearly all BLA PNs independent of tdT-Y2R fluorescence, consistent with presynaptic Y2Rs on somatostatin interneurons mediating the above effects. However, only tdT-Y2R-expressing PNs responded to [ahx5-24]NPY with an enhancement of the IsAHP Ultimately, increased PN excitability via acute Y2R activation likely correlates with enhanced BLA output, consistent with reported Y2R-mediated anxiogenesis. Furthermore, we demonstrate the following: (1) a novel mechanism whereby activity-independent GABA release can powerfully dampen BLA neuronal excitability via postsynaptic GABABRs; and (2) that this tonic inhibition can be interrupted by neuromodulation, here by NPY via Y2Rs.SIGNIFICANCE STATEMENT Within the BLA, NPY is potently anxiolytic. However, selective activation of NPY2 receptors (Y2Rs) increases anxiety by an unknown mechanism. We show that activation of BLA Y2Rs decreases tonic GABA release onto BLA principal neurons, probably from Y2R-expressing somatostatin interneurons, some of which coexpress NPY. This increases principal neuron excitability by reducing GABAB receptor (GABABR)-mediated activation of G-protein-coupled, inwardly rectifying K+ currents. Tonic, Y2R-sensitive GABABR currents unexpectedly persisted in the absence of action potential firing, revealing, to our knowledge, the first report of substantial, activity-independent GABABR activation. Ultimately, we provide a plausible explanation for Y2R-mediated anxiogenesis in vivo and describe a novel and modulatable means of damping neuronal excitability.


Subject(s)
Action Potentials/drug effects , Basolateral Nuclear Complex/drug effects , Neural Inhibition/drug effects , Neurons/drug effects , Receptors, Neuropeptide Y/agonists , Animals , Female , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice , Miniature Postsynaptic Potentials/drug effects , Rats , Rats, Sprague-Dawley
4.
J Neurosci ; 39(23): 4448-4460, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30936241

ABSTRACT

Striatal output pathways are known to play a crucial role in the control of movement. One possible component for shaping the synaptic output of striatal neuron is the glutamatergic input that originates from cortex and thalamus. Although reports focusing on quantifying glutamatergic-induced morphological changes in striatum exist, the role of glutamatergic input in regulating striatal function remains poorly understood. Using primary neurons from newborn mice of either sex in a reduced two-neuron microcircuit culture system, we examined whether glutamatergic input modulates the output of striatal neurons. We found that glutamatergic input enhanced striatal inhibition in vitro With a glutamatergic partner from either cortex or thalamus, we attributed this potentiation to an increase in the size of quantal IPSC, suggesting a strengthening of the postsynaptic response to GABAergic signaling. Additionally, a differential effect of cortical and thalamic innervation onto striatal GABAergic neurons output was revealed. We observed that cortical, but not thalamic input, enhanced the number of releasable GABAergic synaptic vesicles and morphological synapses. Importantly, these alterations were reverted by blockade of neuronal activity and glutamate receptors, as well as disruption of BDNF-TrkB signaling. Together, our data indicate, for first time, that GABAergic synapse formation in corticostriatal pairs depends on two parallel, but potentially intersecting, signaling pathways that involve glutamate receptor activation in striatal neurons, as well as BDNF signaling. Understanding how cortical and thalamic inputs refine striatal output will pave the way toward dissecting basal ganglia activity in both physiological and pathological conditions.SIGNIFICANCE STATEMENT Striatal GABAergic microcircuits are critical for motor function. However, the mechanisms controlling striatal output, particularly at the level of synaptic strength, are unclear. Using two-neuron culture system, we quantified the synaptic output of individual striatal GABAergic neurons paired with a glutamatergic partner and studied the influence of the excitatory connections that are known to be interregionally formed in vivo We found that glutamatergic input potentiated striatal inhibitory output, potentially involving an increased feedback and/or feedforward inhibition. Moreover, distinct components of glutamatergic innervation, such as firing activity or release of neurotrophic factors were shown to be required for the glutamatergic-induced phenotype. Investigation, therefore, of two-neuron in vitro microcircuits could be a powerful tool to explore synaptic mechanisms or disease pathophysiology.


Subject(s)
Corpus Striatum/physiology , GABAergic Neurons/physiology , Glutamic Acid/physiology , Synapses/physiology , gamma-Aminobutyric Acid/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Antibodies, Neutralizing/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/physiology , Cells, Cultured , Cerebral Cortex/cytology , Corpus Striatum/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Female , GABAergic Neurons/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Protein-Tyrosine Kinases/physiology , Quinoxalines/pharmacology , Recombinant Proteins/pharmacology , Synaptic Vesicles/physiology , Tetrodotoxin/pharmacology , Thalamus/cytology
5.
J Neurosci ; 39(23): 4422-4433, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30926749

ABSTRACT

The discovery of a G-protein-coupled receptor for lactate named hydroxycarboxylic acid receptor 1 (HCAR1) in neurons has pointed to additional nonmetabolic effects of lactate for regulating neuronal network activity. In this study, we characterized the intracellular pathways engaged by HCAR1 activation, using mouse primary cortical neurons from wild-type (WT) and HCAR1 knock-out (KO) mice from both sexes. Using whole-cell patch clamp, we found that the activation of HCAR1 with 3-chloro-5-hydroxybenzoic acid (3Cl-HBA) decreased miniature EPSC frequency, increased paired-pulse ratio, decreased firing frequency, and modulated membrane intrinsic properties. Using fast calcium imaging, we show that HCAR1 agonists 3,5-dihydroxybenzoic acid, 3Cl-HBA, and lactate decreased by 40% spontaneous calcium spiking activity of primary cortical neurons from WT but not from HCAR1 KO mice. Notably, in neurons lacking HCAR1, the basal activity was increased compared with WT. HCAR1 mediates its effect in neurons through a Giα-protein. We observed that the adenylyl cyclase-cAMP-protein kinase A axis is involved in HCAR1 downmodulation of neuronal activity. We found that HCAR1 interacts with adenosine A1, GABAB, and α2A-adrenergic receptors, through a mechanism involving both its Giα and Gißγ subunits, resulting in a complex modulation of neuronal network activity. We conclude that HCAR1 activation in neurons causes a downmodulation of neuronal activity through presynaptic mechanisms and by reducing neuronal excitability. HCAR1 activation engages both Giα and Gißγ intracellular pathways to functionally interact with other Gi-coupled receptors for the fine tuning of neuronal activity.SIGNIFICANCE STATEMENT Expression of the lactate receptor hydroxycarboxylic acid receptor 1 (HCAR1) was recently described in neurons. Here, we describe the physiological role of this G-protein-coupled receptor (GPCR) and its activation in neurons, providing information on its expression and mechanism of action. We dissected out the intracellular pathway through which HCAR1 activation tunes down neuronal network activity. For the first time, we provide evidence for the functional cross talk of HCAR1 with other GPCRs, such as GABAB, adenosine A1- and α2A-adrenergic receptors. These results set HCAR1 as a new player for the regulation of neuronal network activity acting in concert with other established receptors. Thus, HCAR1 represents a novel therapeutic target for pathologies characterized by network hyperexcitability dysfunction, such as epilepsy.


Subject(s)
Heterotrimeric GTP-Binding Proteins/physiology , Lactates/metabolism , Nerve Tissue Proteins/physiology , Neurons/physiology , Receptors, G-Protein-Coupled/physiology , Action Potentials , Animals , Calcium Signaling/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cyclic AMP/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/drug effects , Primary Cell Culture , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Second Messenger Systems/drug effects
6.
J Neurosci ; 39(25): 4874-4888, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30992373

ABSTRACT

Surgical ovariectomy has been shown to reduce spine density in hippocampal CA1 pyramidal cells of rodents, and this reduction is reversed by 17ß-estradiol (E2) treatment in a model of human estrogen replacement therapy. Here, we report reduction of spine density in apical dendrites of layer 5 pyramidal neurons of several neocortical regions that is reversed by subsequent E2 treatment in ovariectomized (OVX) female Thy1M-EGFP mice. We also found that OVX-associated reduction of spine density in somatosensory cortex was accompanied by a reduction in miniature EPSC (mEPSC) frequency (but not mIPSC frequency), indicating a change in functional synapses. OVX-associated spine loss in somatosensory cortex was also rescued by an agonist of the G-protein-linked estrogen receptor (GPER) but not by agonists of the classic estrogen receptors ERα/ERß, whereas the opposite selectivity was found in area CA1. Acute treatment of neocortical slices with E2 also rescued the OVX-associated reduction in mEPSC frequency, which could be mimicked by a GPER agonist and abolished by a GPER antagonist. Time-lapse in vivo two-photon imaging showed that OVX-associated reduction in spine density is achieved by both an increase in spine loss rate and a decrease in spine gain rate and that subsequent rescue by E2 reversed both of these processes. Crucially, the spines added after E2 rescue were no more likely to reappear at or nearby the sites of pre-OVX spines than those in control mice treated with vehicle. Thus, a model of estrogen replacement therapy, although restoring spine density and dynamics, does not entirely restore functional connectivity.SIGNIFICANCE STATEMENT Estrogen replacement therapy following menopause or surgical removal of the ovaries is a widespread medical practice, yet little is known about the consequences of such treatment for cells in the brain. Here, we show that estrogen replacement reverses some of the effects of surgical removal of the ovaries on the structure and function of brain cells in the mouse. Yet, importantly, the fine wiring of the brain is not returned to the presurgery state by estrogen treatment, suggesting lasting functional consequences.


Subject(s)
Dendritic Spines/drug effects , Estradiol/pharmacology , Neocortex/drug effects , Pyramidal Cells/drug effects , Animals , Dendritic Spines/metabolism , Estrogens/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , Mice , Miniature Postsynaptic Potentials/drug effects , Neocortex/cytology , Neocortex/metabolism , Ovariectomy , Pyramidal Cells/cytology , Pyramidal Cells/metabolism
7.
Biochem Biophys Res Commun ; 532(3): 489-495, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32892950

ABSTRACT

The GABAA receptor (GABAAR) is the main inhibitory receptor in the adult mammalian brain. GABAAR function is dependent on its expression, distribution, and the chloride (Cl-) transmembrane gradient, which is determined by the potassium-chloride cotransporter 2 (KCC2) in the adult brain. KCC2 and GABAAR are downregulated in an activity-dependent manner during seizure induction. Functionally, KCC2 and GABAAR are closely related membrane proteins which modulate GABAergic inhibition. However, it remains unclear how their downregulation during seizure induction is coordinated. This study aimed to assess this interaction. Our results revealed that KCC2 and GABAAR were simultaneously downregulated in both in vivo and in vitro seizure models induced by the convulsant cyclothazide (CTZ), which was at least partly due to structural coupling in hippocampal neuronal membranes. Immunohistochemistry revealed colocalization of gephyrin with KCC2 and co-immunoprecipitation exhibited a direct coupling between GABAAR α1-subunit and KCC2 protein in hippocampal cell membranes. KCC2 specific short hairpin RNA (KCC2-shRNA) was employed to specifically reduce the expression of KCC2 in cultured hippocampal neurons. This resulted in a significant reduction in KCC2-independent GABAergic miniature inhibitory post-synaptic current (mIPSC) amplitude in shKCC2-transfected neurons. Further, pre-treatment with furosemide, a KCC2 inhibitor, during CTZ stimulation followed by washout significantly prevented convulsant stimulation-induced membrane KCC2 downregulation and significantly attenuated GABAAR downregulation concomitant with recovery of suppressed KCC2-independent GABAergic mIPSC amplitude. Our results suggest that the coordinated downregulation of KCC2 and GABAAR during seizure induction exerts a strong functional impact on GABAAR, highlighting an important regulatory mechanism in epilepsy.


Subject(s)
Receptors, GABA-A/metabolism , Seizures/metabolism , Symporters/metabolism , Animals , Benzothiadiazines/toxicity , Brain/drug effects , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Male , Miniature Postsynaptic Potentials/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Seizures/chemically induced , Symporters/antagonists & inhibitors , Symporters/genetics , K Cl- Cotransporters
8.
Pharmacol Res ; 161: 105128, 2020 11.
Article in English | MEDLINE | ID: mdl-32805354

ABSTRACT

Cannabidiol (CBD) is a major phytocannabinoid in Cannabis sativa. CBD is being increasingly reported as a clinical treatment for neurological diseases. Febrile seizure is one of the most common diseases in children with limited therapeutic options. We investigated possible therapeutic effects of CBD on febrile seizures and the underlying mechanism. Use of a hyperthermia-induced seizures model revealed that CBD significantly prolonged seizure latency and reduced the severity of thermally-induced seizures. Hippocampal neuronal excitability was significantly decreased by CBD. Further, CBD significantly reduced the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated evoked excitatory postsynaptic currents (eEPSCs) and the amplitude and frequency of miniature EPSCs (mEPSCs). Furthermore, CBD significantly accelerated deactivation in GluA1 and GluA2 subunits. Interestingly, CBD slowed receptor recovery from desensitization of GluA1, but not GluA2. These effects on kinetics were even more prominent when AMPAR was co-expressed with γ-8, the high expression isoform 8 of transmembrane AMPAR regulated protein (TARPγ8) in the hippocampus. The inhibitory effects of CBD on AMPAR depended on its interaction with the distal N-terminal domain of GluA1/GluA2. CBD inhibited AMPAR activity and reduced hippocampal neuronal excitability, thereby improving the symptoms of febrile seizure in mice. The putative binding site of CBD in the N-terminal domain of GluA1/GluA2 may be a drug target for allosteric gating modulation of AMPAR.


Subject(s)
Anticonvulsants/pharmacology , Brain Waves/drug effects , CA1 Region, Hippocampal/drug effects , Cannabidiol/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hyperthermia/complications , Receptors, AMPA/antagonists & inhibitors , Seizures, Febrile/prevention & control , Animals , Anticonvulsants/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Cannabidiol/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/metabolism , Excitatory Postsynaptic Potentials/drug effects , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Kinetics , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Models, Molecular , Protein Binding , Reaction Time/drug effects , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Seizures, Febrile/etiology , Seizures, Febrile/metabolism , Seizures, Febrile/physiopathology
9.
Addict Biol ; 25(3): e12748, 2020 05.
Article in English | MEDLINE | ID: mdl-30963693

ABSTRACT

The United States is experiencing an opioid crisis imposing enormous fiscal and societal costs and driving the staggering overdose death rate. While prescription opioid analgesics are essential for treating acute pain, cessation of use in individuals with a physical dependence induces an aversive withdrawal syndrome that promotes continued drug use to alleviate/avoid these symptoms. Additionally, repeated bouts of withdrawal often lead to an increased propensity for relapse. Understanding the neurobiology underlying withdrawal is essential for providing novel treatment options to alleviate physiological and affective components accompanying the cessation of opiate use. Here, we administered morphine and precipitated withdrawal with naloxone to investigate behavioral and cellular responses in C57BL/6J male and female mice. Following 3 days of administration, both male and female mice demonstrated sensitized withdrawal symptoms. Since the bed nucleus of the stria terminalis (BNST) plays a role in mediating withdrawal-associated behaviors, we examined plastic changes in inhibitory synaptic transmission within this structure 24 hours following the final precipitated withdrawal. In male mice, morphine withdrawal increased spontaneous GABAergic signaling compared with controls. In contrast, morphine withdrawal decreased spontaneous GABAergic signaling in female mice. Intriguingly, these opposing GABAergic effects were contingent upon activity-dependent dynamics within the ex vivo slice. Our findings suggest that male and female mice exhibit some divergent cellular responses in the BNST following morphine withdrawal, and alterations in BNST inhibitory signaling may contribute to the expression of behaviors following opioid withdrawal.


Subject(s)
Analgesics, Opioid/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neural Inhibition/drug effects , Septal Nuclei/drug effects , Substance Withdrawal Syndrome/physiopathology , Synaptic Transmission/drug effects , Animals , Female , Male , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Morphine Dependence , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Septal Nuclei/cytology , Septal Nuclei/metabolism , Septal Nuclei/physiopathology , Substance Withdrawal Syndrome/etiology , gamma-Aminobutyric Acid/metabolism
10.
Pharmacology ; 103(1-2): 68-75, 2019.
Article in English | MEDLINE | ID: mdl-30513516

ABSTRACT

The Medial Habenular (MHb) and the Lateral Habenular nuclei are 2 main parts of the habenular complex (Hb). Recent studies showed that MHb plays an important role in memory, and in the expression of ErbB4. However, the expression of MHb ErbB4 receptor and its role in fear memory is not well understood. In this study, western blotting and quantitative real-time polymerase chain reaction were used to assess the protein and mRNA levels of ErbB4 in the process of contextual fear conditioning. A pharmacological approach was used to block and stimulate the ErbB4 receptor. Contextual fear conditioning tests induced a significant increase on the expression of ErbB4 at various times in the Hb and the MHb. Moreover, the blockade and stimulation of MHb ErbB4 receptors did not affect the fear formation but impaired and improved the contextual-dependent fear expression. Furthermore, in vitro electrophysiological recordings showed that the blockade of the MHb ErbB4 receptor reduced the presynaptic gamma-amino butyric acid release. ErbB4 is a susceptible gene for schizophrenia and the above findings may provide new insights into the mechanisms of fear-related responses.


Subject(s)
Fear/physiology , Habenula/metabolism , Memory/physiology , Receptor, ErbB-4/metabolism , Animals , Behavior Rating Scale , Conditioning, Classical , Fear/psychology , Freezing Reaction, Cataleptic/drug effects , Habenula/drug effects , Habenula/physiology , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Neuregulin-1/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-4/agonists , Receptor, ErbB-4/antagonists & inhibitors , Receptor, ErbB-4/genetics , Tyrphostins/pharmacology
11.
Pharmacology ; 103(1-2): 76-81, 2019.
Article in English | MEDLINE | ID: mdl-30517935

ABSTRACT

Dextromethorphan (DEX) presynaptically decreases glutamatergic transmission in second-order neurons of the nucleus tractus solitarius (TS). To clarify the inhibitory mechanism of DEX, the present study examined the interaction of DEX with cAMP. The effects of DEX on miniature and TS-evoked excitatory postsynaptic currents (mEPSCs and eEPSCs) were recorded under activation of the cAMP-dependent pathway using the brainstem slices. An increase in cAMP by forskolin counteracted the inhibitory effect of DEX on mEPSCs. Eight-Bromo-cAMP and N-ethylmaleimide also attenuated the DEX effect. However, forskolin had negligible effects on the DEX-induced inhibition of eEPSCs. This suggests that DEX decreases spontaneous glutamate release by inhibiting the cAMP-dependent pathway and synchronous release by another unknown mechanism.


Subject(s)
Cyclic AMP/metabolism , Dextromethorphan/pharmacology , Glutamates/metabolism , Neurons/drug effects , Solitary Nucleus/drug effects , Solitary Nucleus/physiology , Synaptic Transmission/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Colforsin/pharmacology , Ethylmaleimide/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Guinea Pigs , Male , Miniature Postsynaptic Potentials/drug effects , Neurons/metabolism , Neurons/physiology , Solitary Nucleus/metabolism , Synaptic Transmission/physiology
12.
Biochim Biophys Acta Biomembr ; 1860(5): 1000-1007, 2018 May.
Article in English | MEDLINE | ID: mdl-29317196

ABSTRACT

The formerly widely used broad-spectrum biocide triclosan (TCS) has now become a subject of special concern due to its accumulation in the environment and emerging diverse toxicity. Despite the common opinion that TCS is an uncoupler of oxidative phosphorylation in mitochondria, there have been so far no studies of protonophoric activity of this biocide on artificial bilayer lipid membranes (BLM). Yet only few works have indicated the relationship between TCS impacts on mitochondria and nerve cell functioning. Here, we for the first time report data on a high protonophoric activity of TCS on planar BLM. TCS proved to be a more effective protonophore on planar BLM, than classical uncouplers. Correlation between a strong depolarizing effect of TCS on bacterial membranes and its bactericidal action on Bacillus subtilis might imply substantial contribution of TCS protonophoric activity to its antimicrobial efficacy. Protonophoric activity of TCS, monitored by proton-dependent mitochondrial swelling, resulted in Ca2+ efflux from mitochondria. A comparison of TCS effects on molluscan neurons with those of conventional mitochondrial uncouplers allowed us to ascribe the TCS-induced neuronal depolarization and suppression of excitability to the consequences of mitochondrial deenergization. Also similar to the action of common uncouplers, TCS caused a pronounced increase in frequency of miniature end-plate potentials at neuromuscular junctions. Thus, the TCS-induced mitochondrial uncoupling could alter neuronal function through distortion of Ca2+ homeostasis.


Subject(s)
Calcium/metabolism , Membrane Potentials/drug effects , Miniature Postsynaptic Potentials/drug effects , Mitochondria, Liver/drug effects , Protons , Triclosan/pharmacology , Action Potentials/drug effects , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Lymnaea , Membrane Potentials/physiology , Mice , Miniature Postsynaptic Potentials/physiology , Mitochondria, Liver/metabolism , Mitochondrial Swelling/drug effects , Mitochondrial Swelling/physiology , Oxidative Phosphorylation/drug effects , Rats , Uncoupling Agents/pharmacology
13.
J Neurosci Res ; 96(6): 1066-1079, 2018 06.
Article in English | MEDLINE | ID: mdl-29436006

ABSTRACT

At the mouse neuromuscular junction, adenosine triphosphate (ATP) is co-released with the neurotransmitter acetylcholine (ACh), and once in the synaptic cleft, it is hydrolyzed to adenosine. Both ATP/adenosine diphosphate (ADP) and adenosine modulate ACh secretion by activating presynaptic P2Y13 and A1 , A2A , and A3 receptors, respectively. To elucidate the action of endogenous purines on K+ -dependent ACh release, we studied the effect of purinergic receptor antagonists on miniature end-plate potential (MEPP) frequency in phrenic diaphragm preparations. At 10 mM K+ , the P2Y13 antagonist N-[2-(methylthio)ethyl]-2-[3,3,3-trifluoropropyl]thio-5'-adenylic acid, monoanhydride with (dichloromethylene)bis[phosphonic acid], tetrasodium salt (AR-C69931MX) increased asynchronous ACh secretion while the A1 , A3 , and A2A antagonists 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), (3-Ethyl-5-benzyl-2-methyl-4-phenylethynyl-6-phenyl-1, 4-(±)-dihydropyridine-3,5-, dicarboxylate (MRS-1191), and 2-(2-Furanyl)-7-(2-phenylethyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (SCH-58261) did not modify neurosecretion. The inhibition of equilibrative adenosine transporters by S-(p-nitrobenzyl)-6-thioinosine provoked a reduction of 10 mM K+ -evoked ACh release, suggesting that the adenosine generated from ATP is being removed from the synaptic space by the transporters. At 15 and 20 mM K+ , endogenous ATP/ADP and adenosine bind to inhibitory P2Y13 and A1 and A3 receptors since AR-C69931MX, DPCPX, and MRS-1191 increased MEPP frequency. Similar results were obtained when the generation of adenosine was prevented by using the ecto-5'-nucleotidase inhibitor α,ß-methyleneadenosine 5'-diphosphate sodium salt. SCH-58261 only reduced neurosecretion at 20 mM K+ , suggesting that more adenosine is needed to activate excitatory A2A receptors. At high K+ concentration, the equilibrative transporters appear to be saturated allowing the accumulation of adenosine in the synaptic cleft. In conclusion, when motor nerve terminals are depolarized by increasing K+ concentrations, the ATP/ADP and adenosine endogenously generated are able to modulate ACh secretion by sequential activation of different purinergic receptors.


Subject(s)
Acetylcholine/metabolism , Miniature Postsynaptic Potentials/drug effects , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Potassium/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Purines/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Female , Male , Mice , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Receptors, Purinergic P1/metabolism , Thionucleotides/pharmacology , Triazoles/pharmacology
14.
Article in English | MEDLINE | ID: mdl-29290056

ABSTRACT

Endogenous monoamine 5-hydroxytryptamine (5-HT, serotonin) is a phylogenetically ancient neurotransmitter present in vertebrates. The functions of 5-HT in central nervous system are intensively studied; however, the presynaptic effects of 5-HT in frog spinal motoneurons are practically unexplored. We have previously shown that 5-HT decreases the frequency of glycinergic miniature inhibitory postsynaptic potentials (mIPSPs), but does not affect the frequency of GABAergic mIPSPs and increases the frequency of glutamatergic postsynaptic potentials. In the present study, using pharmacological methods and intracellular recordings in motoneurons from an adult frog's isolated spinal cord, we aimed to identify the 5-HT receptor subtype responsible for inhibiting the release of glycine. Аn agonist of 5-HT1A and 5-HT7 receptors, 8-OH-DPAT, and a selective agonist of 5-HT2 receptors, α-Ме-5-НТ, did not show any significant effect on inhibitory transmission, indicating that 5-HT1A, 5-HT2, and 5-HT7 receptors are not involved in the modulation of glycine release in the adult frog spinal cord. An agonist of 5-HT1B/D receptors sumatriptan decreased the frequency (but not the amplitude) of glycinergic mIPSPs similar to 5-HT. An antagonist of 5-HT1,2 receptors, methysergide, abolished the effect of sumatriptan. Together our results suggest that 5-HT inhibits the release of glycine by activation of 5-HT1B/D receptors.


Subject(s)
Glycine/metabolism , Motor Neurons/metabolism , Neural Inhibition/physiology , Presynaptic Terminals/metabolism , Receptors, Serotonin, 5-HT1/metabolism , Spinal Cord/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Methysergide/pharmacology , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Motor Neurons/drug effects , Neural Inhibition/drug effects , Presynaptic Terminals/drug effects , Rana ridibunda , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT2/metabolism , Serotonin/metabolism , Serotonin Agents/pharmacology , Tissue Culture Techniques
15.
Epilepsia ; 59(2): 333-344, 2018 02.
Article in English | MEDLINE | ID: mdl-29315524

ABSTRACT

OBJECTIVE: Phenobarbital is the most commonly utilized drug for the treatment of neonatal seizures. The use of phenobarbital continues despite growing evidence that it exerts suboptimal seizure control and is associated with long-term alterations in brain structure, function, and behavior. Alterations following neonatal phenobarbital exposure include acute induction of neuronal apoptosis, disruption of synaptic development in the striatum, and a host of behavioral deficits. These behavioral deficits include those in learning and memory mediated by the hippocampus. However, the synaptic changes caused by acute exposure to phenobarbital that lead to lasting effects on brain function and behavior remain understudied. METHODS: Postnatal day (P)7 rat pups were treated with phenobarbital (75 mg/kg) or saline. On P13-14 or P29-37, acute slices were prepared and whole-cell patch-clamp recordings were made from CA1 pyramidal neurons. RESULTS: At P14 we found an increase in miniature inhibitory postsynaptic current (mIPSC) frequency in the phenobarbital-exposed as compared to the saline-exposed group. In addition to this change in mIPSC frequency, the phenobarbital group displayed larger bicuculline-sensitive tonic currents, decreased capacitance and membrane time constant, and a surprising persistence of giant depolarizing potentials. At P29+, the frequency of mIPSCs in the saline-exposed group had increased significantly from the frequency at P14, typical of normal synaptic development; at this age the phenobarbital-exposed group displayed a lower mIPSC frequency than did the control group. Spontaneous inhibitory postsynaptic current (sIPSC) frequency was unaffected at either P14 or P29+. SIGNIFICANCE: These neurophysiological alterations following phenobarbital exposure provide a potential mechanism by which acute phenobarbital exposure can have a long-lasting impact on brain development and behavior.


Subject(s)
CA1 Region, Hippocampal/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/drug effects , Phenobarbital/pharmacology , Pyramidal Cells/drug effects , Synapses/drug effects , Animals , Animals, Newborn , CA1 Region, Hippocampal/cytology , Electric Capacitance , Patch-Clamp Techniques , Pyramidal Cells/metabolism , Rats , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
16.
J Neurosci Res ; 95(7): 1391-1401, 2017 07.
Article in English | MEDLINE | ID: mdl-27770577

ABSTRACT

There is some evidence that glutamate (Glu) acts as a signaling molecule at vertebrate neuromuscular junctions where acetylcholine (ACh) serves as a neurotransmitter. In this study, performed on the cutaneous pectoris muscle of the frog Rana ridibunda, Glu receptor mechanisms that modulate ACh release processes were analyzed. Electrophysiological experiments showed that Glu reduces both spontaneous and evoked quantal secretion of ACh and synchronizes its release in response to electrical stimulation. Quisqualate, an agonist of ionotropic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors and metabotropic Group I mGlu receptors, also exerted Glu-like inhibitory effects on the secretion of ACh but had no effect on the kinetics of quantal release. Quisqualate's inhibitory effect did not occur when a blocker of Group I mGlu receptors (LY 367385) or an inhibitor of phospholipase C (U73122) was present. An increase in the degree of synchrony of ACh quantal release, such as that produced by Glu, was obtained after application of N-methyl-D-aspartic acid (NMDA). The presence of Group I mGlu and NMDA receptors in the neuromuscular synapse was confirmed by immunocytochemistry. Thus, the data suggest that both metabotropic Group I mGlu receptors and ionotropic NMDA receptors are present at the neuromuscular synapse of amphibians, and that the activation of these receptors initiates different mechanisms for the regulation of ACh release from motor nerve terminals. © 2016 Wiley Periodicals, Inc.


Subject(s)
Acetylcholine/metabolism , Miniature Postsynaptic Potentials/physiology , Neuromuscular Junction/metabolism , Receptors, Ionotropic Glutamate/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Miniature Postsynaptic Potentials/drug effects , Neuromuscular Junction/drug effects , Organ Culture Techniques , Rana ridibunda , Receptors, Ionotropic Glutamate/agonists , Receptors, Ionotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors
17.
Synapse ; 71(11)2017 11.
Article in English | MEDLINE | ID: mdl-28857293

ABSTRACT

It has been known that Ca2+ plays an essential role in mediating different modes of neurotransmitter release via different sensing mechanisms. Synaptotagmin 1, 2, and 9 were found to act as the Ca2+ sensors for synchronous release and synaptotagmin 7 and Doc-2 were proposed as the Ca2+ sensors for asynchronous release. Comparatively, the Ca2+ sensor for spontaneous release remains a mystery. At the Calyx of Held synapse, the Ca2+ sensor for spontaneous release was found not identical to the sensor for synchronous release, synaptotagmin 2. As Ca2+ sensors have different sensitivity to Sr2+ and Ca2+ and induce significantly different rate of vesicle release, Sr2+ is traditionally used as a tool to examine the intrinsic properties of different Ca2+ sensors. Here, we employed cell-attached patch recording and presynaptic/postsynaptic whole-cell recording at the Calyx of Held synapses of synaptotagmin 2 knock-out mice to assay the Sr2+ and Ca2+ influx into the nerve terminal at resting potential and observed the effects of Ca2+ and Sr2+ on spontaneous neurotransmitter release. We found that the dwell time of single voltage gated Ca2+ channel opening increased around threefold for Sr2+ than Ca2+ with the channel conductance unchanged; the divalent cation sensing machinery in regulating spontaneous release has much lower sensitivity to Sr2+ than Ca2+ . Thus, our study reveals some of the intrinsic properties of Ca2+ sensor(s) of spontaneous transmitter release and provided an insight into the underlying mechanisms.


Subject(s)
Brain Stem/metabolism , Strontium/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , Animals , Auditory Pathways/drug effects , Auditory Pathways/metabolism , Brain Stem/drug effects , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Cations, Divalent/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Strontium/administration & dosage , Synapses/drug effects , Synaptic Vesicles/drug effects , Synaptotagmin II/deficiency , Synaptotagmin II/genetics , Tissue Culture Techniques
18.
Cereb Cortex ; 26(3): 1081-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25452570

ABSTRACT

Adenosine is an endogenous neuromodulator that decreases excitability of hippocampal circuits activating membrane-bound metabotropic A1 receptor (A1R). The presynaptic inhibitory action of adenosine A1R in glutamatergic synapses is well documented, but its influence on inhibitory GABAergic transmission is poorly known. We report that GABAA receptor (GABAAR)-mediated tonic, but not phasic, transmission is suppressed by A1R in hippocampal neurons. Adenosine A1R activation strongly inhibits GABAAR agonist (muscimol)-evoked currents in Cornu Ammonis 1 (CA1) pyramidal neurons and in a specific subpopulation of interneurons expressing axonal cannabinoid receptor type 1. In addition, A1R suppresses tonic GABAAR currents measured in the presence of elevated ambient GABA as well as in naïve slices. The inhibition of GABAergic currents involves both protein kinase A (PKA) and protein kinase C (PKC) signaling pathways and decreases GABAAR δ-subunit expression. On the contrary, no A1R-mediated modulation was detected in phasic inhibitory postsynaptic currents evoked either by afferent electrical stimulation or by spontaneous quantal release. The results show that A1R modulates extrasynaptic rather than synaptic GABAAR-mediated signaling, and that this modulation selectively occurs in hippocampal pyramidal neurons and in a specific subpopulation of inhibitory interneurons. We conclude that modulation of tonic GABAAR signaling by adenosine A1R in specific neuron types may regulate neuronal gain and excitability in the hippocampus.


Subject(s)
CA1 Region, Hippocampal/physiology , Interneurons/physiology , Pyramidal Cells/physiology , Receptor, Adenosine A1/metabolism , Receptors, GABA-A/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Immunoblotting , Immunohistochemistry , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/cytology , Interneurons/drug effects , Male , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Protein Kinase C/metabolism , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats, Wistar , Tissue Culture Techniques
19.
J Neurosci ; 35(36): 12404-11, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26354909

ABSTRACT

The ζ-inhibitory peptide (ZIP) is considered a candidate inhibitor of the atypical protein kinase Mζ (PKMζ). ZIP has been shown to reverse established LTP and disrupt several forms of long-term memory. However, recent studies have challenged the specificity of ZIP, as it was reported to exert its effect also in PKMζ knock-out mice. These results raise the question of what are the targets of ZIP that may underlie its effect on LTP and memory. Here we report that ZIP as well as its inactive analog, scrambled ZIP, induced a dose-dependent increase in spontaneous activity of neurons in dissociated cultures of rat hippocampus. This was followed by a sustained elevation of intracellular calcium concentration ([Ca(2+)]i) which could not be blocked by conventional channel blockers. Furthermore, ZIP caused an increase in frequency of mEPSCs followed by an increase in membrane noise in patch-clamped neurons both in culture and in acute brain slices. Finally, at 5-10 µM, ZIP-induced excitotoxic death of the cultured neurons. Together, our results suggest that the potential contribution of cellular toxicity should be taken into account in interpretation of ZIP's effects on neuronal and behavioral plasticity. Significance statement: The ζ-inhibitory peptide (ZIP) is considered a candidate inhibitor of the atypical protein kinase Mζ (PKMζ). ZIP has been shown to reverse established LTP and disrupt several forms of long-term memory. Here we report that ZIP as well as its inactive analog, scrambled ZIP, induced a dose-dependent increase in spontaneous activity of neurons in dissociated cultures and brain slices of rat hippocampus. Furthermore, ZIP caused a dose- and time-dependent neuronal death in the dissociated cultures. These findings impact on the assumption that ZIP erases memory due to specific inhibition of PKMz.


Subject(s)
Excitatory Postsynaptic Potentials/drug effects , Hippocampus/metabolism , Lipopeptides/toxicity , Miniature Postsynaptic Potentials/drug effects , Neurons/drug effects , Protein Kinase C/antagonists & inhibitors , Animals , Calcium/metabolism , Cell-Penetrating Peptides , Cells, Cultured , Hippocampus/cytology , Hippocampus/physiology , Male , Neurons/metabolism , Neurons/physiology , Protein Kinase C/metabolism , Rats , Rats, Wistar
20.
J Neurophysiol ; 116(3): 1498-506, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27385796

ABSTRACT

The role of central regulatory circuits in modulating diabetes-associated glucose dysregulation has only recently been under rigorous investigation. One brain region of interest is the dorsal motor nucleus of the vagus (DMV), which contains preganglionic parasympathetic motor neurons that regulate subdiaphragmatic visceral function. Previous research has demonstrated that glutamatergic and GABAergic neurotransmission are independently remodeled after chronic hyperglycemia/hypoinsulinemia. However, glutamatergic circuitry within the dorsal brain stem impinges on GABAergic regulation of the DMV. The present study investigated the role of glutamatergic neurotransmission in synaptic GABAergic control of DMV neurons after streptozotocin (STZ)-induced hyperglycemia/hypoinsulinemia by using electrophysiological recordings in vitro. The frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) was elevated in DMV neurons from STZ-treated mice. The effect was abolished in the presence of the ionotropic glutamate receptor blocker kynurenic acid or the sodium channel blocker tetrodotoxin, suggesting that after STZ-induced hyperglycemia/hypoinsulinemia, increased glutamatergic receptor activity occurs at a soma-dendritic location on local GABA neurons projecting to the DMV. Although sIPSCs in DMV neurons normally demonstrated considerable amplitude variability, this variability was significantly increased after STZ-induced hyperglycemia/hypoinsulinemia. The elevated amplitude variability was not related to changes in quantal release, but rather correlated with significantly elevated frequency of sIPSCs in these mice. Taken together, these findings suggest that GABAergic regulation of central vagal circuitry responsible for the regulation of energy homeostasis undergoes complex functional reorganization after several days of hyperglycemia/hypoinsulinemia, including both glutamate-dependent and -independent forms of plasticity.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Vagus Nerve/physiopathology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Kynurenic Acid/pharmacology , Male , Mice , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neurons/drug effects , Patch-Clamp Techniques , Receptors, Glutamate/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Tissue Culture Techniques , Vagus Nerve/drug effects , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL