1
|
Jafari-Sabet M, Amiri S, Sheibani M, Fatahi N, Aghamiri H. Cross state-dependent memory retrieval between tramadol and ethanol: involvement of dorsal hippocampal GABAA receptors. Psychopharmacology (Berl) 2024; 241:139-152. [PMID: 37758936 DOI: 10.1007/s00213-023-06469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
RATIONALE Tramadol and ethanol, as psychoactive agents, are often abused. Discovering the molecular pathways of drug-induced memory creation may contribute to preventing drug addiction and relapse. OBJECTIVE The tramadol- and ethanol-induced state-dependent memory (SDM) and cross-SDM retrieval between tramadol and ethanol were examined in this study. Moreover, because of the confirmed involvement of GABAA receptors and GABAergic neurotransmission in memory retrieval impairment, we assessed cross-SDM retrieval between tramadol and ethanol with a specific emphasis on the role of the GABAA receptors. The first hypothesis of this study was the presence of cross-SDM between tramadol and ethanol, and the second hypothesis was related to possible role of GABAA receptors in memory retrieval impairment within the dorsal hippocampus. The cannulae were inserted into the hippocampal CA1 area of NMRI mice, and a step-down inhibitory avoidance test was used to evaluate state dependence and memory recovery. RESULTS The post-training and/or pre-test administration of tramadol (2.5 and 5 mg/kg, i.p.) and/or ethanol (0.5 and 1 g/kg, i.p.) induced amnesia, which was restored after the administration of the drugs 24 h later during the pre-test period, proposing ethanol and tramadol SDM. The pre-test injection of ethanol (0.25 and 0.5 g/kg, i.p.) with tramadol at an ineffective dose (1.25 mg/kg) enhanced tramadol SDM. Moreover, tramadol injection (1.25 and 2.5 mg/kg) with ethanol at the ineffective dose (0.25 g/kg) promoted ethanol SDM. Furthermore, the pre-test intra-CA1 injection of bicuculline (0.0625, 0.125, and 0.25 μg/mouse), a GABAA receptor antagonist, 5 min before the injection of tramadol (5 mg/kg) or ethanol (1 g/kg) inhibited tramadol- and ethanol-induced SDM dose-dependently. CONCLUSION The findings strongly confirmed cross-SDM between tramadol and ethanol and the critical role of dorsal hippocampal GABAA receptors in the cross-SDM between tramadol and ethanol.
Collapse
Affiliation(s)
- Majid Jafari-Sabet
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Shiva Amiri
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Navid Fatahi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Helia Aghamiri
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Bi-directional modulation of hyperpolarization-activated cation currents (I h) by ethanol in rat hippocampal CA3 pyramidal neurons. Neuropharmacology 2023; 227:109423. [PMID: 36690323 DOI: 10.1016/j.neuropharm.2023.109423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
It is widely acknowledged that ethanol (EtOH) can alter many neuronal functions, including synaptic signaling, firing discharge, and membrane excitability, through its interaction with multiple membrane proteins and intracellular pathways. Previous work has demonstrated that EtOH enhances the firing rate of hippocampal GABAergic interneurons and thus the presynaptic GABA release at CA1 and CA3 inhibitory synapses through a positive modulation of the hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels. Activation of HCN channels produce an inward current, commonly called Ih, which plays an essential role in generating/regulating specific neuronal activities in GABAergic interneurons and principal glutamatergic pyramidal neurons such as those in the CA3 subregion. Since the direct effect of EtOH on HCN channels expressed in CA3 pyramidal neurons was not thoroughly elucidated, we investigated the possible interaction between EtOH and HCN channels and the impact on excitability and postsynaptic integration of these neurons. Patch-clamp recordings were performed in single CA3 pyramidal neurons from acute male rat coronal hippocampal slices. Our results show that EtOH modulates HCN-mediated Ih in a concentration-dependent and bi-directional manner, with a positive modulation at lower (20 mM) and an inhibitory action at higher (60-80 mM) concentrations. The modulation of Ih by EtOH was mimicked by forskolin, antagonized by different drugs that selectively interfere with the AC/cAMP/PKA intracellular pathway, as well as by the selective HCN inhibitor ZD7288. Altogether, these data further support the evidence that HCN channels may represent an important molecular target through which EtOH may regulate neuronal activity.
Collapse
|
3
|
Mira RG, Lira M, Tapia-Rojas C, Rebolledo DL, Quintanilla RA, Cerpa W. Effect of Alcohol on Hippocampal-Dependent Plasticity and Behavior: Role of Glutamatergic Synaptic Transmission. Front Behav Neurosci 2020; 13:288. [PMID: 32038190 PMCID: PMC6993074 DOI: 10.3389/fnbeh.2019.00288] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
Problematic alcohol drinking and alcohol dependence are an increasing health problem worldwide. Alcohol abuse is responsible for approximately 5% of the total deaths in the world, but addictive consumption of it has a substantial impact on neurological and memory disabilities throughout the population. One of the better-studied brain areas involved in cognitive functions is the hippocampus, which is also an essential brain region targeted by ethanol. Accumulated evidence in several rodent models has shown that ethanol treatment produces cognitive impairment in hippocampal-dependent tasks. These adverse effects may be related to the fact that ethanol impairs the cellular and synaptic plasticity mechanisms, including adverse changes in neuronal morphology, spine architecture, neuronal communication, and finally an increase in neuronal death. There is evidence that the damage that occurs in the different brain structures is varied according to the stage of development during which the subjects are exposed to ethanol, and even much earlier exposure to it would cause damage in the adult stage. Studies on the cellular and cognitive deficiencies produced by alcohol in the brain are needed in order to search for new strategies to reduce alcohol neuronal toxicity and to understand its consequences on memory and cognitive performance with emphasis on the crucial stages of development, including prenatal events to adulthood.
Collapse
Affiliation(s)
- Rodrigo G Mira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile
| | - Matias Lira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Providencia, Chile
| | - Daniela L Rebolledo
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Escuela de Obstetricia y Puericultura and Centro Integrativo de Biología y Química Aplicada (CIBQA), Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Providencia, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Neurobiology of Aging, Universidad San Sebastián, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
4
|
Winkler P, Luhmann HJ, Kilb W. Taurine potentiates the anticonvulsive effect of the GABA A agonist muscimol and pentobarbital in the immature mouse hippocampus. Epilepsia 2019; 60:464-474. [PMID: 30682212 DOI: 10.1111/epi.14651] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The high incidence of epileptic seizures in neonates and their frequent refractoriness to pharmacologic therapies require identification of new therapeutical options. Therefore, we investigated whether the modulatory effect of taurine on γ-aminobutyric acid (GABA)A receptors can enhance the anticonvulsive potential of the GABAA receptor agonist muscimol and of the barbiturate pentobarbital. METHODS We performed field potential recordings in in toto hippocampus preparations of immature (postnatal days 4-7) C57Bl/6 mouse pups. Spontaneous epileptiform activity was induced by the continuous presence of the potassium channel blocker 4-aminopyridine and the glycinergic antagonist strychnine in Mg2+ -free solutions. RESULTS Bath application of 0.1 μmol/L muscimol increases the occurrence of recurrent epileptiform discharges, whereas they are significantly attenuated in a dose-dependent manner by muscimol in concentrations between 0.5 and 5 μmol/L. Taurine at concentrations between 0.1 and 0.5 mmol/L induces a proconvulsive effect, but upon coapplication, it significantly augments the anticonvulsive effect of moderate muscimol doses (0.5-1 μmol/L). In addition, the anticonvulsive effect of 100 and 200 μmol/L pentobarbital is increased significantly in the presence of 0.5 μmol/L taurine. SIGNIFICANCE These observations demonstrate that taurine can indeed enhance the anticonvulsive effects of muscimol and pentobarbital, suggesting that taurine may act as a positive modulator on GABAA receptors. Thus, interfering with the modulatory taurine binding site of GABAA receptors or the interstitial taurine concentration may provide new therapeutical options for anticonvulsive therapies in neonates.
Collapse
Affiliation(s)
- Paula Winkler
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
5
|
Subramanian S, Reichard RA, Stevenson HS, Schwartz ZM, Parsley KP, Zahm DS. Lateral preoptic and ventral pallidal roles in locomotion and other movements. Brain Struct Funct 2018; 223:2907-2924. [PMID: 29700637 PMCID: PMC5997555 DOI: 10.1007/s00429-018-1669-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022]
Abstract
The lateral preoptic area (LPO) and ventral pallidum (VP) are structurally and functionally distinct territories in the subcommissural basal forebrain. It was recently shown that unilateral infusion of the GABAA receptor antagonist, bicuculline, into the LPO strongly invigorates exploratory locomotion, whereas bicuculline infused unilaterally into the VP has a negligible locomotor effect, but when infused bilaterally, produces vigorous, abnormal pivoting and gnawing movements and compulsive ingestion. This study was done to further characterize these responses. We observed that bilateral LPO infusions of bicuculline activate exploratory locomotion only slightly more potently than unilateral infusions and that unilateral and bilateral LPO injections of the GABAA receptor agonist muscimol potently suppress basal locomotion, but only modestly inhibit locomotion invigorated by amphetamine. In contrast, unilateral infusions of muscimol into the VP affect basal and amphetamine-elicited locomotion negligibly, but bilateral VP muscimol infusions profoundly suppress both. Locomotor activation elicited from the LPO by bicuculline was inhibited modestly and profoundly by blockade of dopamine D2 and D1 receptors, respectively, but was not entirely abolished even under combined blockade of dopamine D1 and D2 receptors. That is, infusing the LPO with bic caused instances of near normal, even if sporadic, invigoration of locomotion in the presence of saturating dopamine receptor blockade, indicating that LPO can stimulate locomotion in the absence of dopamine signaling. Pivoting following bilateral VP bicuculline infusions was unaffected by dopamine D2 receptor blockade, but was completely suppressed by D1 receptor blockade. The present results are discussed in a context of neuroanatomical and functional organization underlying exploratory locomotion and adaptive movements.
Collapse
Affiliation(s)
- Suriya Subramanian
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA
| | - Rhett A Reichard
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA
| | - Hunter S Stevenson
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA
| | - Zachary M Schwartz
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA
| | - Kenneth P Parsley
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA
| | - Daniel S Zahm
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Saint Louis, MO, 63104, USA.
| |
Collapse
|
6
|
Silvestre de Ferron B, Vilpoux C, Kervern M, Robert A, Antol J, Naassila M, Pierrefiche O. Increase of KCC2 in hippocampal synaptic plasticity disturbances after perinatal ethanol exposure. Addict Biol 2017; 22:1870-1882. [PMID: 27778437 DOI: 10.1111/adb.12465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/13/2016] [Accepted: 09/26/2016] [Indexed: 11/28/2022]
Abstract
Low to moderate perinatal ethanol exposure (PEE) may have disastrous consequences for the central nervous system resulting notably in permanent cognitive deficits. Learning and memory are mediated in the hippocampus by long-term potentiation (LTP) and long term depression (LTD), two forms of synaptic plasticity. PEE decreases LTP but also abnormally facilitates LTD (Kervern et al. ) through a presently unknown mechanism. We studied in rat hippocampus slice, the involvement of the chloride co-transporters NKCC1 and KCC2, in the role of GABAA inhibitions in facilitated LTD after moderate PEE. After PEE and in contrast to control slices, facilitated LTD in CA1 field was reduced by the GABAA receptor antagonist bicuculline with no changes in sensitivity to bicuculline and in GABA and benzodiazepine binding sites. Also, sensitivity to diazepam was unaltered, whereas aberrant LTD was blocked. Immunohistochemistry and protein analysis demonstrated an increase in KCC2 protein level at cell membrane in CA1 after PEE with no change in NKCC1 expression. Specifically, both monomeric and dimeric forms of KCC2 were increased in CA1. Bumetanide (10-100 μM), a dose-dependent blocker of NKCC1 and KCC2, or VU0240551 (10 μM) a specific antagonist of KCC2, corrected the enhanced LTD and interestingly bumetanide also restored the lower LTP after PEE. These results demonstrate for the first time an upregulation of the KCC2 co-transporter expression after moderate PEE associated with disturbances in GABAergic neurotransmission modulating bidirectional synaptic plasticity in the hippocampus. Importantly, bumetanide compensated deficits in both LTP and LTD, revealing its potential therapeutic properties.
Collapse
Affiliation(s)
- Benoît Silvestre de Ferron
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Catherine Vilpoux
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Myriam Kervern
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Alexandre Robert
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Johan Antol
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Mickael Naassila
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| | - Olivier Pierrefiche
- INSERM ERI-24, GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé CHU-Sud, Université Picardie Jules Verne, Amiens, France
| |
Collapse
|
7
|
Wakita M, Shoudai K, Oyama Y, Akaike N. 4,5-Dichloro-2-octyl-4-isothiazolin-3-one (DCOIT) modifies synaptic transmission in hippocampal CA3 neurons of rats. CHEMOSPHERE 2017; 184:337-346. [PMID: 28605704 DOI: 10.1016/j.chemosphere.2017.05.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/23/2017] [Accepted: 05/28/2017] [Indexed: 06/07/2023]
Abstract
4,5-Dichloro-2-octyl-4-isothiazolin-3-one (DCOIT) is an alternative to organotin antifoulants, such as tributyltin and triphenyltin. Since DCOIT is found in harbors, bays, and coastal areas worldwide, this chemical compound may have some impacts on ecosystems. To determine whether DCOIT possesses neurotoxic activity by modifying synaptic transmission, we examined the effects of DCOIT on synaptic transmission in a 'synaptic bouton' preparation of rat brain. DCOIT at concentrations of 0.03-1 μM increased the amplitudes of evoked synaptic currents mediated by GABA and glutamate, while it reduced the amplitudes of these currents at 3-10 μM. However, the currents elicited by exogenous applications of GABA and glutamate were not affected by DCOIT. DCOIT at 1-10 μM increased the frequency of spontaneous synaptic currents mediated by GABA. It also increased the frequency of glutamate-mediated spontaneous currents at0.3-10 μM. The frequencies of miniature synaptic currents mediated by GABA and glutamate, observed in the presence of tetrodotoxin under external Ca2+-free conditions, were increased by 10 μM DCOIT. With the repetitive applications of DCOIT, the frequency of miniature synaptic currents mediated by glutamate was not increased by the second and third applications of DCOIT. Voltage-dependent Ca2+ channels were not affected by DCOIT, but DCOIT slowed the inactivation of voltage-dependent Na+ channels. These results suggest that DCOIT increases Ca2+ release from intracellular Ca2+ stores, resulting in the facilitation of both action potential-dependent and spontaneous neurotransmission, possibly leading to neurotoxicity.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto, 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, Kumamoto, 861-5598, Japan.
| | - Kiyomitsu Shoudai
- Research Division for Life Science, Kumamoto Health Science University, Kumamoto, 861-5598, Japan
| | - Yasuo Oyama
- Laboratory of Cellular Signaling, Faculty of Biosciences and Bioindustry, Tokushima University, Tokushima, 770-8513, Japan.
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto, 860-8518, Japan; Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan.
| |
Collapse
|
8
|
Ondo W. Essential Tremor: What We Can Learn from Current Pharmacotherapy. Tremor Other Hyperkinet Mov (N Y) 2016; 6:356. [PMID: 26989572 PMCID: PMC4790207 DOI: 10.7916/d8k35tc3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/17/2015] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND The pathophysiology of essential tremor, especially at the cellular level, is poorly understood. Although no drug has been specifically designed to treat essential tremor, several medications improve tremor, and others worsen it. Studying the mechanism of actions of these medications can help our understanding of tremor pathophysiology and contribute to future rational drug design. METHODS We reviewed literature, concentrating on mechanisms of action, of various medications that mitigate tremor. RESULTS Many medications have multiple mechanisms of actions, making simple correlations difficult. Medications that increase the duration of opening of gamma-aminobutyric acid (GABA)-A receptors are most consistently associated with tremor improvement. Interestingly, drugs that increase GABA availability have not been associated with improved tremor. Other mechanisms possibly associated with tremor improvement include antagonism of alpha-2 delta subunits associated with calcium channels, inhibition of carbonic anhydrase, and inhibition of the synaptic vesicle protein 2A. Drugs that block voltage-gaited sodium channels do not affect tremor. The ideal beta-adrenergic blocker requires B2 affinity (non-cardiac selective), has no sympathomimetic properties, does not require membrane stabilization properties, and may benefit from good central nervous system penetration. DISCUSSION To date, serendipitous observations have provided most of our understanding of tremor cellular physiology. Based on similarities to currently effective drugs or rational approximations and inferences, several currently available agents should be considered for tremor trials.
Collapse
Affiliation(s)
- William Ondo
- Methodist Neurological Institute, Houston, TX, USA
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
9
|
Wakita M, Kotani N, Akaike N. Effects of propofol on glycinergic neurotransmission in a single spinal nerve synapse preparation. Brain Res 2015; 1631:147-56. [PMID: 26616339 DOI: 10.1016/j.brainres.2015.11.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 12/30/2022]
Abstract
The effects of the intravenous anesthetic, propofol, on glycinergic transmission and on glycine receptor-mediated whole-cell currents (IGly) were examined in the substantia gelatinosa (SG) neuronal cell body, mechanically dissociated from the rat spinal cord. This "synaptic bouton" preparation, which retains functional native nerve endings, allowed us to evaluate glycinergic inhibitory postsynaptic currents (IPSCs) and whole-cell currents in a preparation in which experimental solution could rapidly access synaptic terminals. Synaptic IPSCs were measured as spontaneous (s) and evoked (e) IPSCs. The eIPSCs were elicited by applying paired-pulse focal electrical stimulation, while IGly was evoked by a bath application of glycine. A concentration-dependent enhancement of IGly was observed for ≥10µM propofol. Propofol (≥3µM) significantly increased the frequency of sIPSCs and prolonged the decay time without altering the current amplitude. However, propofol (≥3µM) also significantly increased the mean amplitude of eIPSCs and decreased the failure rate (Rf). A decrease in the paired-pulse ratio (PPR) was noted at higher concentrations (≥10µM). The decay time of eIPSCs was prolonged only at the maximum concentration tested (30µM). Propofol thus acts at both presynaptic glycine release machinery and postsynaptic glycine receptors. At clinically relevant concentrations (<1μM) there was no effect on IGly, sIPSCs or eIPSCs suggesting that at anesthetic doses propofol does not affect inhibitory glycinergic synapses in the spinal cord.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, JyuryoGroup, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya 343-0821, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, JyuryoGroup, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya 343-0821, Japan; Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
10
|
Wakita M, Kotani N, Yamaga T, Akaike N. Nitrous oxide directly inhibits action potential-dependent neurotransmission from single presynaptic boutons adhering to rat hippocampal CA3 neurons. Brain Res Bull 2015; 118:34-45. [PMID: 26343381 DOI: 10.1016/j.brainresbull.2015.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/26/2015] [Accepted: 09/01/2015] [Indexed: 11/16/2022]
Abstract
We evaluated the effects of N2O on synaptic transmission using a preparation of mechanically dissociated rat hippocampal CA3 neurons that allowed assays of single bouton responses evoked from native functional nerve endings. We studied the effects of N2O on GABAA, glutamate, AMPA and NMDA receptor-mediated currents (IGABA, IGlu, IAMPA and INMDA) elicited by exogenous application of GABA, glutamate, (S)-AMPA, and NMDA and spontaneous, miniature, and evoked GABAergic inhibitory and glutamatergic excitatory postsynaptic current (sIPSC, mIPSC, eIPSC, sEPSC, mEPSC and eEPSC) in mechanically dissociated CA3 neurons. eIPSC and eEPSC were evoked by focal electrical stimulation of a single bouton. Administration of 70% N2O altered neither IGABA nor the frequency and amplitude of both sIPSCs and mIPSCs. In contrast, N2O decreased the amplitude of eIPSCs, while increasing failure rates (Rf) and paired-pulse ratios (PPR) in a concentration-dependent manner. On the other hand, N2O decreased IGlu, IAMPA and INMDA. Again N2O did not change the frequency and amplitude of either sEPSCs of mEPSCs. N2O also decreased amplitudes of eEPSCs with increased Rf and PPR. The decay phases of all synaptic responses were unchanged. The present results indicated that N2O inhibits the activation of AMPA/KA and NMDA receptors and also that N2O preferentially depress the action potential-dependent GABA and glutamate releases but had little effects on spontaneous and miniature releases.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama 343-0821, Japan
| | - Toshitaka Yamaga
- Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto 861-5598, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryo Group, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto 861-5598, Japan; Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, Saitama 343-0821, Japan.
| |
Collapse
|
11
|
Wakita M, Nagami H, Takase Y, Nakanishi R, Kotani N, Akaike N. Modifications of excitatory and inhibitory transmission in rat hippocampal pyramidal neurons by acute lithium treatment. Brain Res Bull 2015; 117:39-44. [PMID: 26247839 DOI: 10.1016/j.brainresbull.2015.07.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 07/24/2015] [Accepted: 07/28/2015] [Indexed: 11/25/2022]
Abstract
The acute effects of high-dose Li(+) treatment on glutamatergic and GABAergic transmissions were studied in the "synaptic bouton" preparation of isolated rat hippocampal pyramidal neurons by using focal electrical stimulation. Both action potential-dependent glutamatergic excitatory and GABAergic inhibitory postsynaptic currents (eEPSC and eIPSC, respectively) were dose-dependently inhibited in the external media containing 30-150 mM Li(+), but the sensitivity for Li(+) was greater tendency for eEPSCs than for eIPSCs. When the effects of Li(+) on glutamate or GABAA receptor-mediated whole-cell responses (IGlu and IGABA) elicited by an exogenous application of glutamate or GABA were examined in the postsynaptic soma membrane of CA3 neurons, Li(+) slightly inhibited both IGlu and IGABA at the 150 mM Li(+) concentration. Present results suggest that acute treatment with high concentrations of Li(+) acts preferentially on presynaptic terminals, and that the Li(+)-induced inhibition may be greater for excitatory than for inhibitory transmission.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Juryokai, Kumamoto Kinoh Hospital, 6-8-1, Yamamuro, Kita-ku, Kumamoto 860-8518, Japan
| | - Hideaki Nagami
- Research Division for Clinical Pharmacology, Medical Corporation, Juryokai, Kumamoto Kinoh Hospital, 6-8-1, Yamamuro, Kita-ku, Kumamoto 860-8518, Japan
| | - Yuko Takase
- Research Division for Clinical Pharmacology, Medical Corporation, Juryokai, Kumamoto Kinoh Hospital, 6-8-1, Yamamuro, Kita-ku, Kumamoto 860-8518, Japan
| | - Ryoji Nakanishi
- Research Division for Clinical Pharmacology, Medical Corporation, Juryokai, Kumamoto Kinoh Hospital, 6-8-1, Yamamuro, Kita-ku, Kumamoto 860-8518, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6, Kawarasone, Koshigaya 343-0821, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Juryokai, Kumamoto Kinoh Hospital, 6-8-1, Yamamuro, Kita-ku, Kumamoto 860-8518, Japan; Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6, Kawarasone, Koshigaya 343-0821, Japan; Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
12
|
Wakita M, Kotani N, Shoudai K, Yamaga T, Akaike N. Modulation of inhibitory and excitatory fast neurotransmission in the rat CNS by heavy water (D2O). J Neurophysiol 2015; 114:1109-18. [PMID: 26019316 DOI: 10.1152/jn.00801.2014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/26/2015] [Indexed: 11/22/2022] Open
Abstract
The effects of heavy water (deuterium oxide, D2O) on GABAergic and glutamatergic spontaneous and evoked synaptic transmission were investigated in acute brain slice and isolated "synaptic bouton" preparations of rat hippocampal CA3 neurons. The substitution of D2O for H2O reduced the frequency and amplitude of GABAergic spontaneous inhibitory postsynaptic currents (sIPSCs) in a concentration-dependent manner but had no effect on glutamatergic spontaneous excitatory postsynaptic currents (sEPSCs). In contrast, for evoked synaptic responses in isolated neurons, the amplitude of both inhibitory and excitatory postsynaptic currents (eIPSCs and eEPSCs) was decreased in a concentration-dependent manner. This was associated with increases of synaptic failure rate (Rf) and paired-pulse ratio (PPR). The effect was larger for eIPSCs compared with eEPSCs. These results clearly indicate that D2O acts differently on inhibitory and excitatory neurotransmitter release machinery. Furthermore, D2O significantly suppressed GABAA receptor-mediated whole cell current (IGABA) but did not affect glutamate receptor-mediated whole cell current (IGlu). The combined effects of D2O at both the pre- and postsynaptic sites may explain the greater inhibition of eIPSCs compared with eEPSCs. Finally, D2O did not enhance or otherwise affect the actions of the general anesthetics nitrous oxide and propofol on spontaneous or evoked GABAergic and glutamatergic neurotransmissions, or on IGABA and IGlu. Our results suggest that previously reported effects of D2O to mimic and/or modulate anesthesia potency result from mechanisms other than modulation of GABAergic and glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryo Group, Kumamoto Kinoh Hospital, Kitaku, Kumamoto, Japan; Research Division for Life Science, Kumamoto Health Science University, Kitaku, Kumamoto, Japan; and
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, Koshigaya, Japan
| | - Kiyomitsu Shoudai
- Research Division for Life Science, Kumamoto Health Science University, Kitaku, Kumamoto, Japan; and
| | - Toshitaka Yamaga
- Research Division for Life Science, Kumamoto Health Science University, Kitaku, Kumamoto, Japan; and
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryo Group, Kumamoto Kinoh Hospital, Kitaku, Kumamoto, Japan; Research Division for Life Science, Kumamoto Health Science University, Kitaku, Kumamoto, Japan; and Research Division of Neurophysiology, Kitamoto Hospital, Koshigaya, Japan
| |
Collapse
|
13
|
Wakita M, Kotani N, Akaike N. Tetrodotoxin abruptly blocks excitatory neurotransmission in mammalian CNS. Toxicon 2015; 103:12-8. [PMID: 25959619 DOI: 10.1016/j.toxicon.2015.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/28/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
The present study utilised a 'synaptic bouton' preparation of mechanically isolated rat hippocampal CA3 pyramidal neurons, which permits direct physiological and pharmacological quantitative analyses at the excitatory and inhibitory single synapse level. Evoked excitatory and inhibitory postsynaptic currents (eEPSCs and eIPSCs) were generated by focal paired-pulse electrical stimulation of single boutons. The sensitivity of eEPSC to tetrodotoxin (TTX) was higher than that of the voltage-dependent Na(+) channel whole-cell current (INa) in the postsynaptic CA3 soma membrane. The synaptic transmission was strongly inhibited by 3 nM TTX, at which concentration the INa was hardly suppressed. The IC50 values of eEPSC and INa for TTX were 2.8 and 37.9 nM, respectively, and complete inhibition was 3-10 nM for eEPSC and 1000 nM for INa. On the other hand, both eEPSC and eIPSC were equally and gradually inhibited by decreasing the external Na(+) concentration ([Na]o), which decreases the Na(+)gradient across the cell membrane. The results indicate that TTX at 3-10 nM could block most of voltage-dependent Na(+) channels on presynaptic nerve terminal, resulting in abruptly inhibition of action potential dependent excitatory neurotransmission.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto, 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto, 861-5598, Japan
| | - Naoki Kotani
- Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, 343-0821, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, 6-8-1 Yamamuro, Kitaku, Kumamoto, 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, 325 Izumi-machi, Kitaku, Kumamoto, 861-5598, Japan; Research Division of Neurophysiology, Kitamoto Hospital, 3-7-6 Kawarasone, Koshigaya, 343-0821, Japan.
| |
Collapse
|
14
|
Wakita M, Oyama Y, Takase Y, Akaike N. Modulation of excitatory synaptic transmission in rat hippocampal CA3 neurons by triphenyltin, an environmental pollutant. CHEMOSPHERE 2015; 120:598-607. [PMID: 25462303 DOI: 10.1016/j.chemosphere.2014.09.073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 09/19/2014] [Accepted: 09/22/2014] [Indexed: 06/04/2023]
Abstract
Triphenyltin (TPT) is an organometallic compound that poses a known environmental hazard to some fish and mollusks, as well as mammals. However, its neurotoxic mechanisms in the mammalian brain are still unclear. Thus, we have investigated mechanisms through which TPT modulates glutamatergic synaptic transmission, including spontaneous, miniature, and evoked excitatory postsynaptic currents (sEPSCs, mEPSCs, and eEPSCs respectively), in a rat hippocampal CA3 'synaptic-bouton' preparation. TPT, at environmentally relevant concentrations (30 nM to 1 μM), significantly increased the frequency of sEPSCs and mEPSCs in a concentration-dependent manner, without affecting the currents' amplitudes. The facilitatory effects of TPT on mEPSC frequency were seen even in a Ca(2+)-free external solution containing tetrodotoxin. These effects were further prolonged by adding caffeine, which releases Ca(2+) from intracellular Ca(2+) storage sites. In glutamatergic eEPSCs evoked by paired-pulse stimuli, TPT at concentrations greater than or equal to 100 nM markedly increased the current amplitude by the first pulse and decreased failure rate and pair-pulse ratio. On the other hand, both voltage-dependent Na(+) and Ca(2+) channels were unaffected by submicromolar concentrations of TPT. Overall, these results suggest that TPT, at environmentally relevant concentrations, affects presynaptic transmitter release machinery by directly modulating Ca(2+) storage. Further, findings of this study imply that excitotoxic mechanisms may underlie TPT-induced neuronal damage.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, Kumamoto 861-5598, Japan.
| | - Yasuo Oyama
- Laboratory of Cellular Signaling, Faculty of Integrated Arts and Sciences, University of Tokushima, Tokushima 770-8501, Japan.
| | - Yuko Takase
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto 860-8518, Japan
| | - Norio Akaike
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto 860-8518, Japan; Research Division for Life Science, Kumamoto Health Science University, Kumamoto 861-5598, Japan.
| |
Collapse
|
15
|
Lindsay JH, Glass JD, Amicarelli M, Prosser RA. The mammalian circadian clock in the suprachiasmatic nucleus exhibits rapid tolerance to ethanol in vivo and in vitro. Alcohol Clin Exp Res 2014; 38:760-9. [PMID: 24512529 DOI: 10.1111/acer.12303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/16/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Ethanol (EtOH) triggers cellular adaptations that induce tolerance in many brain areas, including the suprachiasmatic nucleus (SCN), the site of the master circadian clock. EtOH inhibits light-induced phase shifts in the SCN in vivo and glutamate-induced phase shifts in vitro. The in vitro phase shifts develop acute tolerance to EtOH, occurring within minutes of initial exposure, while the in vivo phase shifts exhibit no evidence of chronic tolerance. An intermediate form, rapid tolerance, is not well studied but may predict subsequent chronic tolerance. Here, we investigated rapid tolerance in the SCN clock. METHODS Adult C57BL/6 mice were provided 15% EtOH or water for one 12-hour lights-off period. For in vitro experiments, SCN-containing brain slices were prepared in the morning and treated for 10 minutes with glutamate +/- EtOH the following night. Single-cell neuronal firing rates were recorded extracellularly during the subsequent day to determine SCN clock phase. For in vivo experiments, mice receiving EtOH 24 hours previously were exposed to a 30-minute light pulse immediately preceded by intraperitoneal saline or 2 g/kg EtOH injection. Mice were then placed in constant darkness and their phase-shifting responses measured. RESULTS In vitro, the SCN clock from EtOH-exposed mice exhibited rapid tolerance, with a 10-fold increase in EtOH needed to inhibit glutamate-induced phase shifts. Co-application of brain-derived neurotrophic factor prevented EtOH inhibition, consistent with experiments using EtOH-naïve mice. Rapid tolerance lasts 48 to 96 hours, depending on whether assessing in vitro phase advances or phase delays. Similarly, in vivo, prior EtOH consumption prevented EtOH's acute blockade of photic phase delays. Finally, immunoblot experiments showed no changes in SCN glutamate receptor subunit (NR2B) expression or phosphorylation in response to rapid tolerance induction. CONCLUSIONS The SCN circadian clock develops rapid tolerance to EtOH as assessed both in vivo and in vitro, and the tolerance lasts for several days. These data demonstrate the utility of the circadian system as a model for investigating cellular mechanisms through which EtOH acts in the brain.
Collapse
Affiliation(s)
- Jonathan H Lindsay
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee
| | | | | | | |
Collapse
|
16
|
Wakita M, Kotani N, Kogure K, Akaike N. Inhibition of excitatory synaptic transmission in hippocampal neurons by levetiracetam involves Zn²⁺-dependent GABA type A receptor-mediated presynaptic modulation. J Pharmacol Exp Ther 2014; 348:246-59. [PMID: 24259680 DOI: 10.1124/jpet.113.208751] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Levetiracetam (LEV) is an antiepileptic drug with a unique but as yet not fully resolved mechanism of action. Therefore, by use of a simplified rat-isolated nerve-bouton preparation, we have investigated how LEV modulates glutamatergic transmission from mossy fiber terminals to hippocampal CA3 neurons. Action potential-evoked excitatory postsynaptic currents (eEPSCs) were recorded using a conventional whole-cell patch-clamp recording configuration in voltage-clamp mode. The antiepileptic drug phenytoin decreased glutamatergic eEPSCs in a concentration-dependent fashion by inhibiting voltage-dependent Na⁺ and Ca²⁺ channel currents. In contrast, LEV had no effect on eEPSCs or voltage-dependent Na⁺ or Ca²⁺ channel currents. Activation of presynaptic GABA type A (GABA(A)) receptors by muscimol induced presynaptic inhibition of eEPSCs, resulting from depolarization block. Low concentrations of Zn²⁺, which had no effect on eEPSCs, voltage-dependent Na⁺ or Ca²⁺ channel currents, or glutamate receptor-mediated whole cell currents, reduced the muscimol-induced presynaptic inhibition. LEV applied in the continuous presence of 1 µM muscimol and 1 µM Zn²⁺ reversed this Zn²⁺ modulation on eEPSCs. The antagonizing effect of LEV on Zn²⁺-induced presynaptic GABA(A) receptor inhibition was also observed with the Zn²⁺ chelators Ca-EDTA and RhodZin-3. Our results clearly show that LEV removes the Zn²⁺-induced suppression of GABA(A)-mediated presynaptic inhibition, resulting in a presynaptic decrease in glutamate-mediated excitatory transmission. Our results provide a novel mechanism by which LEV may inhibit neuronal activity.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto, Japan (M.W., N.A.); Research Division for Life Science, Kumamoto Health Science University, Kumamoto, Japan (M.W., N.A.); Research Division of Neurophysiology, Kitamoto Hospital, Koshigaya, Japan (N.K., N.A); and Kogure Medical Clinic, Chouseikai Medical Corporation, Fukaya City, Saitama, Japan (K.K.)
| | | | | | | |
Collapse
|
17
|
Teng SX, Molina PE. Acute alcohol intoxication prolongs neuroinflammation without exacerbating neurobehavioral dysfunction following mild traumatic brain injury. J Neurotrauma 2013; 31:378-86. [PMID: 24050411 DOI: 10.1089/neu.2013.3093] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) represents a leading cause of death and disability among young persons with ∼1.7 million reported cases in the United States annually. Although acute alcohol intoxication (AAI) is frequently present at the time of TBI, conflicting animal and clinical reports have failed to establish whether AAI significantly impacts short-term outcomes after TBI. The objective of this study was to determine whether AAI at the time of TBI aggravates neurobehavioral outcomes and neuroinflammatory sequelae post-TBI. Adult male Sprague-Dawley rats were surgically instrumented with gastric and vascular catheters before a left lateral craniotomy. After recovery, rats received either a primed constant intragastric alcohol infusion (2.5 g/kg+0.3 g/kg/h for 15 h) or isocaloric/isovolumic dextrose infusion followed by a lateral fluid percussion TBI (∼1.4 J, ∼30 ms). TBI induced apnea and a delay in righting reflex. AAI at the time of injury increased the TBI induced delay in righting reflex without altering apnea duration. Neurological and behavioral dysfunction was observed at 6 h and 24 h post-TBI, and this was not exacerbated by AAI. TBI induced a transient upregulation of cortical interleukin (IL)-6 and monocyte chemotactic protein (MCP)-1 mRNA expression at 6 h, which was resolved at 24 h. AAI did not modulate the inflammatory response at 6 h but prevented resolution of inflammation (IL-1, IL-6, tumor necrosis factor-α, and MCP-1 expression) at 24 h post-TBI. AAI at the time of TBI did not delay the recovery of neurological and neurobehavioral function but prevented the resolution of neuroinflammation post-TBI.
Collapse
Affiliation(s)
- Sophie X Teng
- Department of Physiology, Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | | |
Collapse
|
18
|
Korkotian E, Bombela T, Odegova T, Zubov P, Segal M. Ethanol affects network activity in cultured rat hippocampus: mediation by potassium channels. PLoS One 2013; 8:e75988. [PMID: 24260098 PMCID: PMC3829821 DOI: 10.1371/journal.pone.0075988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 08/19/2013] [Indexed: 11/21/2022] Open
Abstract
The effects of ethanol on neuronal network activity were studied in dissociated cultures of rat hippocampus. Exposure to low (0.25–0.5%) ethanol concentrations caused an increase in synchronized network spikes, and a decrease in the duration of individual spikes. Ethanol also caused an increase in rate of miniature spontaneous excitatory postsynaptic currents. Higher concentrations of ethanol eliminated network spikes. These effects were reversible upon wash. The effects of the high, but not the low ethanol were blocked by the GABA antagonist bicuculline. The enhancing action of low ethanol was blocked by apamin, an SK potassium channel antagonist, and mimicked by 1-EBIO, an SK channel opener. It is proposed that in cultured hippocampal networks low concentration of ethanol is associated with SK channel activity, rather than the GABAergic receptor.
Collapse
Affiliation(s)
- Eduard Korkotian
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
- * E-mail:
| | - Tatyana Bombela
- Department of Pharmacognosy, Perm State Pharmaceutical Academy, Perm, Russia
| | - Tatiana Odegova
- Department of Microbiology, Perm State Pharmaceutical Academy, Perm, Russia
| | - Petr Zubov
- Department of Microbiology, Perm State Pharmaceutical Academy, Perm, Russia
| | - Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel
| |
Collapse
|
19
|
Tabakoff B, Hoffman PL. The neurobiology of alcohol consumption and alcoholism: an integrative history. Pharmacol Biochem Behav 2013; 113:20-37. [PMID: 24141171 PMCID: PMC3867277 DOI: 10.1016/j.pbb.2013.10.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 01/17/2023]
Abstract
Studies of the neurobiological predisposition to consume alcohol (ethanol) and to transition to uncontrolled drinking behavior (alcoholism), as well as studies of the effects of alcohol on brain function, started a logarithmic growth phase after the repeal of the 18th Amendment to the United States Constitution. Although the early studies were primitive by current technological standards, they clearly demonstrated the effects of alcohol on brain structure and function, and by the end of the 20th century left little doubt that alcoholism is a "disease" of the brain. This review traces the history of developments in the understanding of ethanol's effects on the most prominent inhibitory and excitatory systems of brain (GABA and glutamate neurotransmission). This neurobiological information is integrated with knowledge of ethanol's actions on other neurotransmitter systems to produce an anatomical and functional map of ethanol's properties. Our intent is limited in scope, but is meant to provide context and integration of the actions of ethanol on the major neurobiologic systems which produce reinforcement for alcohol consumption and changes in brain chemistry that lead to addiction. The developmental history of neurobehavioral theories of the transition from alcohol drinking to alcohol addiction is presented and juxtaposed to the neurobiological findings. Depending on one's point of view, we may, at this point in history, know more, or less, than we think we know about the neurobiology of alcoholism.
Collapse
Affiliation(s)
- Boris Tabakoff
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| | - Paula L. Hoffman
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| |
Collapse
|
20
|
Wakita M, Kotani N, Nonaka K, Shin MC, Akaike N. Effects of propofol on GABAergic and glutamatergic transmission in isolated hippocampal single nerve-synapse preparations. Eur J Pharmacol 2013; 718:63-73. [DOI: 10.1016/j.ejphar.2013.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023]
|
21
|
Potent and direct presynaptic modulation of glycinergic transmission in rat spinal neurons by atrial natriuretic peptide. Brain Res Bull 2013; 99:19-26. [PMID: 24060848 DOI: 10.1016/j.brainresbull.2013.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 09/10/2013] [Accepted: 09/16/2013] [Indexed: 11/19/2022]
Abstract
Atrial and brain natriuretic peptides (ANP and BNP) exist in the central nervous system and modulate neuronal function, although the locus of actions and physiological mechanisms are still unclear. In the present study we used rat spinal sacral dorsal commissural nucleus (SDCN) and hippocampal 'synaptic bouton' preparations, to record both spontaneous and evoked glycinergic inhibitory postsynaptic currents (sIPSCs and eIPSCs) in SDCN neurons, and the evoked excitatory postsynaptic currents (eEPSCs) in hippocampal CA3 neurons. ANP potently and significantly reduced the sIPSC frequency without affecting the amplitude. ANP also potently reduced the eIPSCs amplitude concurrently increasing the failure rate and the paired pulse ratio response. These ANP actions were blocked by anantin, a specific type A natriuretic peptide receptor (NPR-A) antagonist. The results clearly indicate that ANP acts directly on glycinergic presynaptic nerve terminals to inhibit glycine release via presynaptic NPR-A. The ANP effects were not blocked by the membrane permeable cGMP analog (8Br-cGMP) suggesting a transduction mechanisms not simply related to increasing cGMP levels in nerve terminals. BNP did not affect on glycinergic sIPSCs and eIPSCs. Moreover, both ANP and BNP had no effect on glutamatergic EPSCs in hippocampal CA3 neurons. The results indicate a potent and selective presynaptic inhibitory action of ANP on glycinergic transmission in spinal cord sensory circuits.
Collapse
|
22
|
Alkohol induzierte kognitive Dysfunktion. Wien Med Wochenschr 2013; 164:9-14. [DOI: 10.1007/s10354-013-0226-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/25/2013] [Indexed: 01/30/2023]
|
23
|
Iwata S, Wakita M, Shin MC, Fukuda A, Akaike N. Modulation of allopregnanolone on excitatory transmitters release from single glutamatergic terminal. Brain Res Bull 2013; 93:39-46. [DOI: 10.1016/j.brainresbull.2012.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 11/02/2012] [Accepted: 11/05/2012] [Indexed: 10/27/2022]
|
24
|
Jiang L, Gulanski BI, De Feyter HM, Weinzimer SA, Pittman B, Guidone E, Koretski J, Harman S, Petrakis IL, Krystal JH, Mason GF. Increased brain uptake and oxidation of acetate in heavy drinkers. J Clin Invest 2013; 123:1605-14. [PMID: 23478412 DOI: 10.1172/jci65153] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 01/17/2013] [Indexed: 11/17/2022] Open
Abstract
When a person consumes ethanol, the body quickly begins to convert it to acetic acid, which circulates in the blood and can serve as a source of energy for the brain and other organs. This study used 13C magnetic resonance spectroscopy to test whether chronic heavy drinking is associated with greater brain uptake and oxidation of acetic acid, providing a potential metabolic reward or adenosinergic effect as a consequence of drinking. Seven heavy drinkers, who regularly consumed at least 8 drinks per week and at least 4 drinks per day at least once per week, and 7 light drinkers, who consumed fewer than 2 drinks per week were recruited. The subjects were administered [2-13C]acetate for 2 hours and scanned throughout that time with magnetic resonance spectroscopy of the brain to observe natural 13C abundance of N-acetylaspartate (NAA) and the appearance of 13C-labeled glutamate, glutamine, and acetate. Heavy drinkers had approximately 2-fold more brain acetate relative to blood and twice as much labeled glutamate and glutamine. The results show that acetate transport and oxidation are faster in heavy drinkers compared with that in light drinkers. Our finding suggests that a new therapeutic approach to supply acetate during alcohol detoxification may be beneficial.
Collapse
Affiliation(s)
- Lihong Jiang
- Department of Diagnostic Radiology, Yale University, School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Dixon CI, Walker SE, King SL, Stephens DN. Deletion of the gabra2 gene results in hypersensitivity to the acute effects of ethanol but does not alter ethanol self administration. PLoS One 2012; 7:e47135. [PMID: 23115637 PMCID: PMC3480382 DOI: 10.1371/journal.pone.0047135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/10/2012] [Indexed: 12/04/2022] Open
Abstract
Human genetic studies have suggested that polymorphisms of the GABRA2 gene encoding the GABAA α2-subunit are associated with ethanol dependence. Variations in this gene also convey sensitivity to the subjective effects of ethanol, indicating a role in mediating ethanol-related behaviours. We therefore investigated the consequences of deleting the α2-subunit on the ataxic and rewarding properties of ethanol in mice. Ataxic and sedative effects of ethanol were explored in GABAA α2-subunit wildtype (WT) and knockout (KO) mice using a Rotarod apparatus, wire hang and the duration of loss of righting reflex. Following training, KO mice showed shorter latencies to fall than WT littermates under ethanol (2 g/kg i.p.) in both Rotarod and wire hang tests. After administration of ethanol (3.5 g/kg i.p.), KO mice took longer to regain the righting reflex than WT mice. To ensure the acute effects are not due to the gabra2 deletion affecting pharmacokinetics, blood ethanol concentrations were measured at 20 minute intervals after acute administration (2 g/kg i.p.), and did not differ between genotypes. To investigate ethanol’s rewarding properties, WT and KO mice were trained to lever press to receive increasing concentrations of ethanol on an FR4 schedule of reinforcement. Both WT and KO mice self-administered ethanol at similar rates, with no differences in the numbers of reinforcers earned. These data indicate a protective role for α2-subunits, against the acute sedative and ataxic effects of ethanol. However, no change was observed in ethanol self administration, suggesting the rewarding effects of ethanol remain unchanged.
Collapse
Affiliation(s)
- Claire I. Dixon
- School of Psychology, University of Sussex, Brighton, United Kingdom
| | - Sophie E. Walker
- School of Psychology, University of Sussex, Brighton, United Kingdom
| | - Sarah L. King
- School of Psychology, University of Sussex, Brighton, United Kingdom
| | - David N. Stephens
- School of Psychology, University of Sussex, Brighton, United Kingdom
- * E-mail:
| |
Collapse
|