Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.060
Filter
1.
Neurobiol Dis ; 164: 105610, 2022 03.
Article in English | MEDLINE | ID: mdl-34995754

ABSTRACT

Aberrant glucocorticoid signaling via glucocorticoid receptors (GR) plays a critical role in alcohol use disorder (AUD). Acute alcohol withdrawal and protracted abstinence in dependent rats are associated with increased GR signaling and changes in GR-mediated transcriptional activity in the rat central nucleus of the amygdala (CeA). The GR antagonist mifepristone decreases alcohol consumption in dependent rats during acute withdrawal and protracted abstinence. Regulation of CeA synaptic activity by GR is currently unknown. Here, we utilized mifepristone and the selective GR antagonist CORT118335 (both at 10 µM) as pharmacological tools to dissect the role of GR on GABA transmission in male, adult Sprague-Dawley rats using slice electrophysiology. We subjected rats to chronic intermittent alcohol vapor exposure for 5-7 weeks to induce alcohol dependence. A subset of dependent rats subsequently underwent protracted alcohol withdrawal for 2 weeks, and air-exposed rats served as controls. Mifepristone reduced the frequency of pharmacologically-isolated spontaneous inhibitory postsynaptic currents (sIPSC) in the CeA (medial subdivision) without affecting postsynaptic measures in all groups, suggesting decreased GABA release with the largest effect in dependent rats. CORT118335 did not significantly alter GABA transmission in naïve, but decreased sIPSC frequency in dependent rats. Similarly, mifepristone decreased amplitudes of evoked inhibitory postsynaptic potentials only in dependent rats and during protracted withdrawal. Collectively, our study provides insight into regulation of CeA GABAergic synapses by GR. Chronic ethanol enhances the efficiency of mifepristone and CORT118335, thus highlighting the potential of drugs targeting GR as a promising pharmacological avenue for the treatment of AUD.


Subject(s)
Alcoholism/physiopathology , Amygdala/drug effects , GABAergic Neurons/drug effects , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors , Synapses/drug effects , Amygdala/physiopathology , Animals , GABAergic Neurons/physiology , Inhibitory Postsynaptic Potentials/drug effects , Male , Rats , Rats, Sprague-Dawley , Synapses/physiology
2.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34502543

ABSTRACT

To elucidate why naftopidil increases the frequency of spontaneous synaptic currents in only some substantia gelatinosa (SG) neurons, post-hoc analyses were performed. Blind patch-clamp recording was performed using slice preparations of SG neurons from the spinal cords of adult rats. Spontaneous inhibitory and excitatory postsynaptic currents (sIPSCs and sEPSCs, respectively) were recorded. The ratios of the frequency and amplitude of the sIPSCs and sEPSCs following the introduction of naftopidil compared with baseline, and after the application of naftopidil, serotonin (5-HT), and prazosin, compared with noradrenaline (NA) were evaluated. First, the sIPSC analysis indicated that SG neurons reached their full response ratio for NA at 50 µM. Second, they responded to 5-HT (50 µM) with a response ratio similar to that for NA, but prazosin (10 µM) did not change the sEPSCs and sIPSCs. Third, the highest concentration of naftopidil (100 µM) led to two types of response in the SG neurons, which corresponded with the reactions to 5-HT and prazosin. These results indicate that not all neurons were necessarily activated by naftopidil, and that the micturition reflex may be regulated in a sophisticated manner by inhibitory mechanisms in these interneurons.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Neurons/drug effects , Patch-Clamp Techniques/methods , Substantia Gelatinosa/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Naphthalenes/pharmacology , Neurons/physiology , Norepinephrine/pharmacology , Piperazines/pharmacology , Prazosin/pharmacology , Rats, Sprague-Dawley , Serotonin/pharmacology , Substantia Gelatinosa/cytology , Substantia Gelatinosa/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
3.
Mol Pharmacol ; 100(3): 217-223, 2021 09.
Article in English | MEDLINE | ID: mdl-34135098

ABSTRACT

Regulators of G protein signaling (RGS) proteins modulate signaling by G protein-coupled receptors. Using a knock-in transgenic mouse model with a mutation in Gαo that does not bind RGS proteins (RGS-insensitive), we determined the effect of RGS proteins on presynaptic µ opioid receptor (MOR)-mediated inhibition of GABA release in the ventrolateral periaqueductal gray (vlPAG). The MOR agonists [d-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) and met-enkephalin (ME) inhibited evoked inhibitory postsynaptic currents (eIPSCs) in the RGS-insensitive mice compared with wild-type (WT) littermates, respectively. Fentanyl inhibited eIPSCs similarly in both WT and RGS-insensitive mice. There were no differences in opioid agonist inhibition of spontaneous GABA release between the genotypes. To further probe the mechanism underlying these differences between opioid inhibition of evoked and spontaneous GABA release, specific myristoylated Gα peptide inhibitors for Gαo1 and Gαi1-3 that block receptor-G protein interactions were used to test the preference of agonists for MOR-Gα complexes. The Gαo1 inhibitor reduced DAMGO inhibition of eIPSCs, but Gαi1-3 inhibitors had no effect. Both Gαo1 and Gαi1-3 inhibitors separately reduced fentanyl inhibition of eIPSCs but had no effects on ME inhibition. Gαi1-3 inhibitors blocked the inhibitory effects of ME and fentanyl on miniature postsynaptic current (mIPSC) frequency, but both Gαo1 and Gαi1-3 inhibitors were needed to block the effects of DAMGO. Finally, baclofen-mediated inhibition of GABA release is unaffected in the RGS-insensitive mice and in the presence of Gαo1 and Gαi1-3 inhibitor peptides, suggesting that GABAB receptor coupling to G proteins in vlPAG presynaptic terminals is different than MOR coupling. SIGNIFICANCE STATEMENT: Presynaptic µ opioid receptors (MORs) in the ventrolateral periaqueductal gray are critical for opioid analgesia and are negatively regulated by RGS proteins. These data in RGS-insensitive mice provide evidence that MOR agonists differ in preference for Gαo versus Gαi and regulation by RGS proteins in presynaptic terminals, providing a mechanism for functional selectivity between agonists. The results further define important differences in MOR and GABAB receptor coupling to G proteins that could be exploited for new pain therapies.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Presynaptic Terminals/physiology , Receptors, Opioid, mu/physiology , gamma-Aminobutyric Acid/metabolism , Analgesics, Opioid/pharmacology , Animals , Baclofen/pharmacology , Female , GTP-Binding Protein alpha Subunit, Gi2/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Transgenic , Models, Animal , RGS Proteins/metabolism , Receptors, GABA-B/metabolism , Receptors, Opioid, mu/agonists
4.
Mol Neurobiol ; 58(10): 4787-4801, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34173171

ABSTRACT

Glutamate delta-1 receptor (GluD1) is a member of the ionotropic glutamate receptor family expressed at excitatory synapses and functions as a synaptogenic protein by interacting with presynaptic neurexin. We have previously shown that GluD1 plays a role in the maintenance of excitatory synapses in a region-specific manner. Loss of GluD1 leads to reduced excitatory neurotransmission in medium spiny neurons (MSNs) in the dorsal striatum, but not in the ventral striatum (both core and shell of the nucleus accumbens (NAc)). Here, we found that GluD1 loss leads to reduced inhibitory neurotransmission in MSNs of the NAc core as evidenced by a reduction in the miniature inhibitory postsynaptic current frequency and amplitude. Presynaptic effect of GluD1 loss was further supported by an increase in paired pulse ratio of evoked inhibitory responses indicating reduced release probability. Furthermore, analysis of GAD67 puncta indicated a reduction in the number of putative inhibitory terminals. The changes in mIPSC were independent of cannabinoid or dopamine signaling. A role of feed-forward inhibition was tested by selective ablation of GluD1 from PV neurons which produced modest reduction in mIPSCs. Behaviorally, local ablation of GluD1 from NAc led to hypolocomotion and affected anxiety- and depression-like behaviors. When GluD1 was ablated from the dorsal striatum, several behavioral phenotypes were altered in opposite manner compared to GluD1 ablation from NAc. Our findings demonstrate that GluD1 regulates inhibitory neurotransmission in the NAc by a combination of pre- and postsynaptic mechanisms which is critical for motor control and behaviors relevant to neuropsychiatric disorders.


Subject(s)
Anxiety/metabolism , Glutamate Dehydrogenase/biosynthesis , Inhibitory Postsynaptic Potentials/physiology , Neural Inhibition/physiology , Nucleus Accumbens/metabolism , Synaptic Transmission/physiology , Animals , Anxiety/genetics , Excitatory Amino Acid Antagonists/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/genetics , Inhibitory Postsynaptic Potentials/drug effects , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Knockout , Neural Inhibition/drug effects , Nucleus Accumbens/drug effects , Social Interaction/drug effects , Synaptic Transmission/drug effects
5.
Neurosci Lett ; 756: 135950, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33979698

ABSTRACT

The mechanisms of general anaesthetics such as propofol have drawn substantial attention. The effects of propofol on inhibitory postsynaptic currents are not exactly the same in different brain nuclei. Recent studies revealed that the paraventricular thalamic nucleus (PVT) is a critical nucleus modulating wakefulness. However, the effects of propofol on PVT neurons and the mechanisms underlying such effects remain unknown. Here, we performed the whole-cell recording of the PVT neurons in acute brain slices and bath application of propofol. We found that propofol hyperpolarized the membrane potentials of the PVT neurons and suppressed the action potentials induced by step-current injection. Propofol did not affect the spontaneous inhibitory postsynaptic currents (sIPSCs) amplitude or frequency, but prolonged the sIPSCs half-width. Besides, propofol increased miniature inhibitory synaptic currents (mIPSCs) frequency and half-width. Furthermore, propofol could induce GABAA receptors-mediated tonic inhibitory currents dose-dependently. Thus, our results demonstrate that propofol hyperpolarizes PVT neurons by modulating inhibitory currents via GABAA receptors in mice.


Subject(s)
Anesthetics, Intravenous/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Midline Thalamic Nuclei/drug effects , Neural Inhibition/drug effects , Neurons/drug effects , Propofol/pharmacology , Animals , Male , Membrane Potentials/drug effects , Mice , Patch-Clamp Techniques
6.
Neuropharmacology ; 192: 108601, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33971215

ABSTRACT

The dorsolateral bed nucleus of the stria terminalis (BNSTDL) has high expression of oxytocin (OT) receptors (OTR), which were shown to facilitate cued fear. However, the role of OTR in the modulation of BNSTDL activity remains elusive. BNSTDL contains GABA-ergic neurons classified based on intrinsic membrane properties into three types. Using in vitro patch-clamp recordings in male rats, we demonstrate that OT selectively excites and increases spontaneous firing rate of Type I BNSTDL neurons. As a consequence, OT increases the frequency, but not amplitude, of spontaneous inhibitory post-synaptic currents (sIPSCs) selectively in Type II neurons, an effect abolished by OTR antagonist or tetrodotoxin, and reduces spontaneous firing rate in these neurons. These results suggest an indirect effect of OT in Type II neurons, which is mediated via OT-induced increase in firing of Type I interneurons. As Type II BNSTDL neurons were shown projecting to the central amygdala (CeA), we also recorded from retrogradely labeled BNST→CeA neurons and we show that OT increases the frequency of sIPSC in these Type II BNST→CeA output neurons. In contrast, in Type III neurons, OT reduces the amplitude, but not frequency, of both sIPSCs and evoked IPSCs via a postsynaptic mechanism without changing their intrinsic excitability. We present a model of fine-tuned modulation of BNSTDL activity by OT, which selectively excites BNSTDL interneurons and inhibits Type II BNST→CeA output neurons. These results suggest that OTR in the BNST might facilitate cued fear by inhibiting the BNST→CeA neurons.


Subject(s)
Central Amygdaloid Nucleus/drug effects , Excitatory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Oxytocin/pharmacology , Septal Nuclei/drug effects , Animals , Central Amygdaloid Nucleus/physiology , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Interneurons/physiology , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Septal Nuclei/physiology
7.
Neuropharmacology ; 191: 108573, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33945826

ABSTRACT

Currently available antidepressants, such as selective serotonin reuptake inhibitors (SSRIs) and serotonin and norepinephrine reuptake inhibitors (SNRIs), generally require weeks to months to produce a therapeutic response, but the mechanism of action underlying the delayed onset of antidepressant-like action remains to be elucidated. The balance between excitatory glutamatergic pyramidal neurons and inhibitory γ-aminobutyric acid (GABA) interneurons, i.e., the excitation:inhibition functional (E:I) balance, in the medial prefrontal cortex (mPFC) is critical in regulating several behaviors and might play an important mediating role in the mechanism of rapid antidepressant-like action reported by several studies. In the present study, the multichannel electrophysiological technique was used to record the firing activities of pyramidal neurons and interneurons and investigate the effects of a single dose of fluoxetine and ketamine (both 10 mg/kg, i.p.) on the E:I functional balance in the rat mPFC after 90 min or 24 h, and the forced swimming test (FST) was used to evaluate the antidepressant-like effects of fluoxetine and ketamine. The present study also explored the effects of chronic treatment with fluoxetine (10 mg/kg, i.g.) for 7 d or 21 d on the E:I functional balance in the mPFC. The present results suggested that a single dose of ketamine could both significantly increase the firing activities of pyramidal neurons and significantly decrease the firing activities of interneurons in the mPFC and exerted significant antidepressant-like action on the FST after 90 min and 24 h, but fluoxetine had no such effects under the same conditions. However, chronic treatment with fluoxetine for 21 d (but not 7 d) could significantly affect the firing activities of pyramidal neurons and interneurons in the mPFC. Taken together, the present results indicated that rapid regulation of the E:I functional balance in the mPFC might be an important common mechanism of rapid-acting antidepressants and the delayed onset of SSRIs might be partly attributed to their inability to rapidly regulate the E:I functional balance in the mPFC. The present study provided a new entry point to the development of rapid-acting antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Behavior, Animal/drug effects , Excitatory Amino Acid Agents , Fluoxetine/pharmacology , Glutamic Acid , Interneurons/drug effects , Ketamine/pharmacology , Male , Pyramidal Cells/drug effects , Rats , Rats, Inbred WF
8.
Int J Mol Sci ; 22(8)2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33918982

ABSTRACT

Lithium (Li+) salt is widely used as a therapeutic agent for treating neurological and psychiatric disorders. Despite its therapeutic effects on neurological and psychiatric disorders, it can also disturb the neuroendocrine axis in patients under lithium therapy. The hypothalamic area contains GABAergic and glutamatergic neurons and their receptors, which regulate various hypothalamic functions such as the release of neurohormones, control circadian activities. At the neuronal level, several neurotransmitter systems are modulated by lithium exposure. However, the effect of Li+ on hypothalamic neuron excitability and the precise action mechanism involved in such an effect have not been fully understood yet. Therefore, Li+ action on hypothalamic neurons was investigated using a whole-cell patch-clamp technique. In hypothalamic neurons, Li+ increased the GABAergic synaptic activities via action potential independent presynaptic mechanisms. Next, concentration-dependent replacement of Na+ by Li+ in artificial cerebrospinal fluid increased frequencies of GABAergic miniature inhibitory postsynaptic currents without altering their amplitudes. Li+ perfusion induced inward currents in the majority of hypothalamic neurons independent of amino-acids receptor activation. These results suggests that Li+ treatment can directly affect the hypothalamic region of the brain and regulate the release of various neurohormones involved in synchronizing the neuroendocrine axis.


Subject(s)
GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Lithium/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synapses/drug effects , Synapses/metabolism , Animals , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Inhibitory Postsynaptic Potentials/drug effects , Patch-Clamp Techniques , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, Amino Acid/metabolism , Synaptic Transmission/drug effects
9.
Neuropharmacology ; 189: 108530, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741404

ABSTRACT

Adolescents are phenotypically characterized with hyper-sensitivity to stress and inappropriate response to stress-inducing events. Despite behavioral distinctions from adults, investigations of developmental shifts in the function of stress peptide corticotropin-releasing factor (CRF) are generally limited. Rodent models have determined that CRF receptor 1 (CRFR1) activation within the central amygdala is associated with a stress response and induces increased GABAergic synaptic neurotransmission within adult males. To investigate age- and sex-specific function of this system, we performed whole-cell patch clamp electrophysiology in brain slices from naive adolescent (postnatal days (P) 40-49) and adult (>P70) male and female Sprague Dawley rats to assess GABAergic activity in the medial central amygdala (CeM). Our results indicate a dynamic influence of age and sex on neuronal excitability within this region, as well as basal spontaneous and miniature (m) inhibitory post-synaptic currents (IPSCs) in the CeM. In addition to replicating prior findings of CRFR1-regulated increases in mIPSC frequency in adult males, we found that the selective CRFR1 agonist, Stressin-1, attenuated mIPSC frequency in adolescent males, at a concentration that did not produce an effect in adult males. Importantly, this age-specific distinction was absent in females, as Stressin-1 attenuated mIPSC frequency in both adolescent and adult females. Finally, an increase in mIPSC frequency in response to the CRF1R antagonist, NBI 35965, was observed only in the CeM of adult males. Together, these data emphasize the robust influence of age and sex on neurophysiological function of a brain region involved in the production of the stress response.


Subject(s)
Central Amygdaloid Nucleus/metabolism , GABAergic Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/metabolism , Acenaphthenes/pharmacology , Age Factors , Animals , Central Amygdaloid Nucleus/drug effects , Female , GABAergic Neurons/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Sex Factors
10.
Nat Commun ; 12(1): 1216, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619249

ABSTRACT

To design potentially more effective therapies, we need to further understand the mechanisms underlying epilepsy. Here, we uncover the role of Rev-erbα in circadian regulation of epileptic seizures. We first show up-regulation of REV-ERBα/Rev-erbα in brain tissues from patients with epilepsy and a mouse model. Ablation or pharmacological modulation of Rev-erbα in mice decreases the susceptibility to acute and chronic seizures, and abolishes diurnal rhythmicity in seizure severity, whereas activation of Rev-erbα increases the animal susceptibility. Rev-erbα ablation or antagonism also leads to prolonged spontaneous inhibitory postsynaptic currents and elevated frequency in the mouse hippocampus, indicating enhanced GABAergic signaling. We also identify the transporters Slc6a1 and Slc6a11 as regulators of Rev-erbα-mediated clearance of GABA. Mechanistically, Rev-erbα promotes the expressions of Slc6a1 and Slc6a11 through transcriptional repression of E4bp4. Our findings propose Rev-erbα as a regulator of synaptic function at the crosstalk between pathways regulating the circadian clock and epilepsy.


Subject(s)
GABAergic Neurons/metabolism , Gene Expression Regulation , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Seizures/genetics , Acute Disease , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Chronic Disease , Disease Models, Animal , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/physiopathology , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Gene Expression Regulation/drug effects , Hippocampus/pathology , Humans , Inhibitory Postsynaptic Potentials/drug effects , Isoquinolines/pharmacology , Kindling, Neurologic/drug effects , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seizures/pathology , Seizures/physiopathology , Small Molecule Libraries/pharmacology , Thiophenes/pharmacology , gamma-Aminobutyric Acid/metabolism
11.
Neuropharmacology ; 187: 108488, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33556384

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (NHIE) is one of the most prevalent causes of death during the perinatal period. The lack of exposure to oxytocin is associated with NHIE-mediated severe brain injury. However, the underlying mechanism is not fully understood. This study combined immunohistochemistry with electrophysiological recordings of hippocampal CA1 neurons to investigate the role of oxytocin in an in vitro model of hypoxic-ischemic (HI) injury (oxygen and glucose deprivation, OGD) in postnatal day 7-10 rats. Immunohistochemical analysis showed that oxytocin largely reduced the relative intensity of TOPRO-3 staining following OGD in the hippocampal CA1 region. Whole-cell patch-clamp recording revealed that the OGD-induced onset time of anoxic depolarization (AD) was significantly delayed by oxytocin. This protective effect of oxytocin was blocked by pretreatment with [d(CH2)51, Tyr (Me)2, Thr4, Orn8, des-Gly-NH29] vasotocin (dVOT, an oxytocin receptor antagonist) or bicuculline (a GABAA receptor antagonist). Interestingly, oxytocin enhanced inhibitory postsynaptic currents in CA1 pyramidal neurons, which were abolished by tetrodotoxin or dVOT. In contrast, oxytocin had no effect on excitatory postsynaptic currents but induced an inward current in 86% of the pyramidal neurons tested. Taken together, these results demonstrate that oxytocin receptor signaling plays a critical role in attenuating neonatal neural death by facilitating GABAergic transmission, which may help to regulate the excitatory-inhibitory balance in local neuronal networks in NHIE patients.


Subject(s)
CA1 Region, Hippocampal/drug effects , Cell Death/drug effects , Hypoxia-Ischemia, Brain/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxytocics/pharmacology , Oxytocin/pharmacology , Receptors, GABA-A/metabolism , Receptors, Oxytocin/metabolism , Animals , Animals, Newborn , CA1 Region, Hippocampal/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Hypoxia-Ischemia, Brain/pathology , Inhibitory Postsynaptic Potentials/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Rats
12.
Neuropharmacology ; 187: 108492, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33582153

ABSTRACT

In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.


Subject(s)
Amacrine Cells/metabolism , Excitatory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/genetics , Orexin Receptors/genetics , Orexins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Synaptic Transmission/genetics , Amacrine Cells/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Orexin Receptors/metabolism , Orexins/pharmacology , Patch-Clamp Techniques , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Retinal Bipolar Cells/drug effects , Retinal Ganglion Cells/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
13.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540803

ABSTRACT

Distinct from ovarian estradiol, the steroid hormone 17ß-estradiol (E2) is produced in the brain and is involved in numerous functions, particularly acting as a neurosteroid. However, the physiological role of E2 and the mechanism of its effects are not well known. In hippocampal slices, 17ß-estradiol has been found to cause a modest increase in fast glutamatergic transmission; because some of these effects are rapid and acute, they might be mediated by membrane-associated receptors via nongenomic action. Moreover, activation of membrane estrogen receptors can rapidly modulate neuron function in a sex-specific manner. To further investigate the neurological role of E2, we examined the effect of E2, as an estrogen receptor (ER) agonist, on synaptic transmission in slices of the prefrontal cortex (PFC) and hippocampus in both male and female mice. Whole-cell recordings of spontaneous excitatory postsynaptic currents (sEPSC) in the PFC showed that E2 acts as a neuromodulator in glutamatergic transmission in the PFC in both sexes, but often in a cell-specific manner. The sEPSC amplitude and/or frequency responded to E2 in three ways, namely by significantly increasing, decreasing or having no response. Additional experiments using an agonist selective for ERß, diarylpropionitrile (DPN) showed that in males the sEPSC and spontaneous inhibitory postsynaptic currents sIPSC responses were similar to their E2 responses, but in females the estrogen receptor ß (ERß) agonist DPN did not influence excitatory transmission in the PFC. In contrast, in the hippocampus of both sexes E2 potentiated the gluatmatergic synaptic transmission in a subset of hippocampal cells. These data indicate that activation of E2 targeting probably a estrogen subtypes or different downstream signaling affect synaptic transmission in the brain PFC and hippocampus between males versus females mice.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/physiology , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Synaptic Transmission/physiology , Animals , Estrogen Receptor alpha/agonists , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , GABA Agents/pharmacology , Hippocampus/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Kinetics , Male , Mice , Mice, Inbred C57BL , Nitriles/pharmacology , Patch-Clamp Techniques , Prefrontal Cortex/drug effects , Propionates/pharmacology , Sex Characteristics , Synaptic Transmission/drug effects
14.
Epilepsia ; 62(2): 542-556, 2021 02.
Article in English | MEDLINE | ID: mdl-33452820

ABSTRACT

OBJECTIVE: Many antiseizure drugs (ASDs) act on voltage-dependent sodium channels, and the molecular basis of these effects is well established. In contrast, how ASDs act on the level of neuronal networks is much less understood. METHODS: In the present study, we determined the effects of eslicarbazepine (S-Lic) on different types of inhibitory neurons, as well as inhibitory motifs. Experiments were performed in hippocampal slices from both sham-control and chronically epileptic pilocarpine-treated rats. RESULTS: We found that S-Lic causes an unexpected reduction of feed-forward inhibition in the CA1 region at high concentrations (300 µM), but not at lower concentrations (100 µM). Concurrently, 300 but not 100 µM S-Lic significantly reduced maximal firing rates in putative feed-forward interneurons located in the CA1 stratum radiatum of sham-control and epileptic animals. In contrast, feedback inhibition was not inhibited by S-Lic. Instead, application of S-Lic, in contrast to previous data for other drugs like carbamazepine (CBZ), resulted in a lasting potentiation of feedback inhibitory post-synaptic currents (IPSCs) only in epileptic and not in sham-control animals, which persisted after washout of S-Lic. We hypothesized that this plasticity of inhibition might rely on anti-Hebbian potentiation of excitatory feedback inputs onto oriens-lacunosum moleculare (OLM) interneurons, which is dependent on Ca2+ -permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Indeed, we show that blocking Ca2+ -permeable AMPA receptors completely prevents upmodulation of feedback inhibition. SIGNIFICANCE: These results suggest that S-Lic affects inhibitory circuits in the CA1 hippocampal region in unexpected ways. In addition, ASD actions may not be sufficiently explained by acute effects on their target channels, rather, it may be necessary to take plasticity of inhibitory circuits into account.


Subject(s)
Anticonvulsants/pharmacology , CA1 Region, Hippocampal/drug effects , Dibenzazepines/pharmacology , Epilepsy/physiopathology , Interneurons/drug effects , Neural Inhibition/drug effects , Pyramidal Cells/drug effects , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy/chemically induced , Feedback, Physiological/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/metabolism , Long-Term Potentiation , Muscarinic Agonists/toxicity , Neuronal Plasticity , Neurons/drug effects , Neurons/metabolism , Pilocarpine/toxicity , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism
15.
Br J Anaesth ; 126(3): 674-683, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33388140

ABSTRACT

BACKGROUND: Multiple cognitive and psychiatric disorders are associated with an increased tonic inhibitory conductance that is generated by α5 subunit-containing γ-aminobutyric acid type A (α5 GABAA) receptors. Negative allosteric modulators that inhibit α5 GABAA receptors (α5-NAMs) are being developed as treatments for these disorders. The effects of α5-NAMs have been studied on recombinant GABAA receptors expressed in non-neuronal cells; however, no study has compared drug effects on the tonic conductance generated by native GABAA receptors in neurones, which was the goal of this study. METHODS: The effects of five α5-NAMs (basmisanil, Ono-160, L-655,708, α5IA, and MRK-016) on tonic current evoked by a low concentration of GABA were studied using whole-cell recordings in cultured mouse hippocampal neurones. Drug effects on current evoked by a saturating concentration of GABA and on miniature inhibitory postsynaptic currents (mIPSCs) were also examined. RESULTS: The α5-NAMs caused a concentration-dependent decrease in tonic current. The potencies varied as the inhibitory concentration for 50% inhibition (IC50) of basmisanil (127 nM) was significantly higher than those of the other compounds (0.4-0.8 nM). In contrast, the maximal efficacies of the drugs were similar (35.5-51.3% inhibition). The α5-NAMs did not modify current evoked by a saturating GABA concentration or mIPSCs. CONCLUSIONS: Basmisanil was markedly less potent than the other α5-NAMs, an unexpected result based on studies of recombinant α5 GABAA receptors. Studying the effects of α5 GABAA receptor-selective drugs on the tonic inhibitory current in neurones could inform the selection of compounds for future clinical trials.


Subject(s)
Cognitive Dysfunction/drug therapy , GABA-A Receptor Antagonists/pharmacology , Hippocampus/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Neurons/drug effects , Receptors, GABA-A/metabolism , Allosteric Regulation , Animals , Cells, Cultured , Cognition/drug effects , Dose-Response Relationship, Drug , Hippocampus/metabolism , Mice , Neurons/metabolism , Patch-Clamp Techniques
16.
Sci Rep ; 11(1): 1716, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33462326

ABSTRACT

Prenatal ethanol exposure causes a variety of cognitive deficits that have a persistent impact on quality of life, some of which may be explained by ethanol-induced alterations in interneuron function. Studies from several laboratories, including our own, have demonstrated that a single binge-like ethanol exposure during the equivalent to the third trimester of human pregnancy leads to acute apoptosis and long-term loss of interneurons in the rodent retrosplenial cortex (RSC). The RSC is interconnected with the hippocampus, thalamus, and other neocortical regions and plays distinct roles in visuospatial processing and storage, as well as retrieval of hippocampal-dependent episodic memories. Here we used slice electrophysiology to characterize the acute effects of ethanol on GABAergic neurotransmission in the RSC of neonatal mice, as well as the long-term effects of neonatal ethanol exposure on parvalbumin-interneuron mediated neurotransmission in adolescent mice. Mice were exposed to ethanol using vapor inhalation chambers. In postnatal day (P) 7 mouse pups, ethanol unexpectedly failed to potentiate GABAA receptor-mediated synaptic transmission. Binge-like ethanol exposure of P7 mice expressing channel rhodopsin in parvalbumin-positive interneurons enhanced the peak amplitudes, asynchronous activity and total charge, while decreasing the rise-times of optically-evoked GABAA receptor-mediated inhibitory postsynaptic currents in adolescent animals. These effects could partially explain the learning and memory deficits that have been documented in adolescent and young adult mice exposed to ethanol during the third trimester-equivalent developmental period.


Subject(s)
Ethanol/pharmacology , Gyrus Cinguli/drug effects , Interneurons/metabolism , Parvalbumins/metabolism , Animals , Animals, Newborn , Female , Gyrus Cinguli/pathology , Gyrus Cinguli/physiology , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/physiology , Male , Mice , Mice, Transgenic , Pregnancy , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Synaptic Transmission/drug effects
17.
Article in English | MEDLINE | ID: mdl-32818535

ABSTRACT

Soluble amyloid beta (Aß) is believed to contribute to cognitive deficits in the early stages of Alzheimer's disease (AD). Increased soluble Aß1-42 in the hippocampus is closely correlated with spatial learning and memory deficits in AD. Riluzole (RLZ), an FDA-approved drug for amyotrophic lateral sclerosis (ALS), has beneficial effects for AD. However, the mechanism underlying the effects remains unclear. In this study, its neuroprotective effect against soluble Aß1-42-induced spatial cognitive deficits in rats was assessed. We found that intrahippocampal injection of soluble Aß1-42 impaired spatial cognitive function and suppressed long-term potentiation (LTP) of the DG region, which was relevant to soluble Aß1-42-induced shift of the hippocampal excitation/inhibition balance toward excitation. Interestingly, RLZ ameliorated Aß1-42-induced behavioral and LTP impairments through rescuing the soluble Aß1-42-induced excitation/inhibition imbalance. RLZ attenuated Aß1-42-mediated facilitation of excitatory synaptic transmission by facilitating glutamate reuptake and decreasing presynaptic glutamate release. Meanwhile, RLZ attenuated the suppression of inhibitory synaptic transmission caused by Aß1-42 by potentiating postsynaptic GABA receptor function. These results suggest that RLZ exerts a neuroprotective effect against soluble Aß1-42-related spatial cognitive deficits through rescuing the excitation/inhibition imbalance, and it could be a potential therapy for AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Dentate Gyrus/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Peptide Fragments/toxicity , Receptors, GABA , Riluzole/pharmacology , Spatial Memory/drug effects , Animals , Dentate Gyrus/physiology , Glutamic Acid/metabolism , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Injections, Intraventricular , Male , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptors, GABA/physiology , Spatial Memory/physiology
18.
Brain Res ; 1750: 147149, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33035497

ABSTRACT

Menthol, which acts as an agonist for transient receptor potential melastatin 8 (TRPM8), has complex effects on nociceptive transmission, including pain relief and hyperalgesia. Here, we addressed the effects of menthol on spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs, respectively) in medullary dorsal horn neurons, using a whole-cell patch-clamp technique. Menthol significantly increased sEPSC frequency, in a concentration-dependent manner, without affecting current amplitudes. The menthol-induced increase in sEPSC frequency could be completely blocked by AMTB, a TRPM8 antagonist, but was not blocked by HC-030031, a transient receptor potential ankyrin 1 (TRPA1) antagonist. Menthol still increased sEPSC frequency in the presence of Cd2+, a general voltage-gated Ca2+ channel blocker, suggesting that voltage-gated Ca2+ channels are not involved in the menthol-induced increase in sEPSC frequency. However, menthol failed to increase sEPSC frequency in the absence of extracellular Ca2+, suggesting that TRPM8 on primary afferent terminals is Ca2+ permeable. On the other hand, menthol also increased sIPSC frequency, without affecting current amplitudes. The menthol-induced increase in sIPSC frequency could be completely blocked by either AMTB or CNQX, an AMPA/KA receptor antagonist, suggesting that the indirect increase in excitability of inhibitory interneurons may lead to the facilitation of spontaneous GABA and/or glycine release. The present results suggested that menthol exerts analgesic effects, via the enhancement of inhibitory synaptic transmission, through central feed-forward neural circuits within the medullary dorsal horn region.


Subject(s)
Menthol/pharmacology , Posterior Horn Cells/metabolism , Synaptic Transmission/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Female , Inhibitory Postsynaptic Potentials/drug effects , Male , Menthol/metabolism , Patch-Clamp Techniques , Posterior Horn Cells/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/cytology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Synaptic Transmission/physiology , TRPA1 Cation Channel/metabolism , TRPC Cation Channels/metabolism
19.
Mol Neurobiol ; 58(1): 156-169, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32909150

ABSTRACT

Focal cortical dysplasia (FCD) is a major cause for drug-resistant epilepsies. The molecular and cellular mechanisms of epileptogenesis in FCD are still poorly understood. Some studies have suggested that deficiencies of γ-aminobutyric acid (GABA) system may play an important role in type II FCD, but it remains controversial. In order to examine whether and how GABAergic interneurons and synaptic function are affected, we generated a somatic mTOR hyperactivation-based mouse model of type II FCD by in utero electroporation, quantified densities of interneurons in the malformed cortices, and recorded miniature inhibitory postsynaptic currents in dysmorphic neurons. We detected 20-25% reduction of GABAergic interneurons within malformed cortices, independent of cortical regions and cell subtypes but proportionate to the decrease of global neuron counts. GABAergic synaptic transmission from interneurons to mTOR hyperactivated dysmorphic neurons was dramatically disrupted, outweighing the decrease of interneuron counts. Postnatal mTOR inhibition partially rescued these alterations of GABAergic system. We also quantified the expression of GABAA receptor, GABA transporter, and chloridion transporter encoding genes and found that their expression was relatively intact within the malformed cortices. Taken together, these results confirmed that GABAergic interneuron and synapse transmission are disturbed profoundly in an mTOR-dependent manner in type II FCD. Our study suggests that postsynaptic mechanisms independent of interneuron reduction or altered expression of GABA synapse genes might be accountable for the impaired GABAergic neurotransmission in type II FCD as well as other mTOR-related epilepsies.


Subject(s)
GABAergic Neurons/metabolism , Interneurons/metabolism , Malformations of Cortical Development/pathology , Malformations of Cortical Development/physiopathology , Synaptic Transmission , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Disease Models, Animal , GABAergic Neurons/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Malformations of Cortical Development/genetics , Mice, Transgenic , Neocortex/pathology , Sirolimus/pharmacology , Synaptic Transmission/drug effects
20.
Mol Neurobiol ; 58(1): 92-105, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32895785

ABSTRACT

Alcohol use-associated disorders are highly comorbid with anxiety disorders; however, their mechanism remains unknown. The amygdala plays a central role in anxiety. We recently found that 7,8-dihydroxyflavone (7,8-DHF) significantly reduces withdrawal symptoms in a rat model of chronic intermittent alcohol (ethanol) exposure. This study aimed to determine the role of 7,8-DHF in regulating anxiety induced by chronic alcohol exposure and its associated underlying mechanism. Male C57BL/6J mice were exposed to chronic intermittent alcohol for 3 weeks followed by alcohol withdrawal for 12 h with or without 7,8-DHF administered intraperitoneally. All mice were tested using an open field test and elevated plus maze to assess anxiety-like behaviors. Synaptic activity and intrinsic excitability in basal and lateral amygdala (BLA) neurons were assessed using electrophysiological recordings. 7,8-DHF alleviated alcohol-induced anxiety-like behavior and attenuated alcohol-induced enhancement of activities in BLA pyramidal neurons. Furthermore, 7,8-DHF prevented alcohol withdrawal-evoked augmentation of glutamatergic transmission in the amygdala and had no effect on GABAergic transmission in the amygdala, as demonstrated by unaltered frequency and amplitude of spontaneous inhibitory postsynaptic currents. Microinjection of K252a, a tropomyosin-related kinase B (TrkB) antagonist, into the BLA blocked the effects of 7,8-DHF on anxiety-like behavior and neuronal activity in the BLA. Our findings suggest that 7,8-DHF alleviates alcohol-induced anxiety-like behavior induced by chronic alcohol exposure through regulation of glutamate transmission involving TrKB in the BLA.


Subject(s)
Amygdala/enzymology , Anxiety/drug therapy , Anxiety/etiology , Behavior, Animal , Flavones/therapeutic use , Receptor, trkB/metabolism , Animals , Anxiety/enzymology , Behavior, Animal/drug effects , Carbazoles/pharmacology , Disease Models, Animal , Ethanol , Excitatory Postsynaptic Potentials/drug effects , Flavones/pharmacology , Glutamic Acid/metabolism , Indole Alkaloids/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Substance Withdrawal Syndrome/pathology , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...