51
|
Söderpalm B, Lidö HH, Ericson M. The Glycine Receptor-A Functionally Important Primary Brain Target of Ethanol. Alcohol Clin Exp Res 2017; 41:1816-1830. [PMID: 28833225 DOI: 10.1111/acer.13483] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/15/2017] [Indexed: 12/27/2022]
Abstract
Identification of ethanol's (EtOH) primary molecular brain targets and determination of their functional role is an ongoing, important quest. Pentameric ligand-gated ion channels, that is, the nicotinic acetylcholine receptor, the γ-aminobutyric acid type A receptor, the 5-hydroxytryptamine3 , and the glycine receptor (GlyR), are such targets. Here, aspects of the structure and function of these receptors and EtOH's interaction with them are briefly reviewed, with special emphasis on the GlyR and the importance of this receptor and its ligands for EtOH pharmacology. It is suggested that GlyRs are involved in (i) the dopamine-activating effect of EtOH, (ii) regulating EtOH intake, and (iii) the relapse preventing effect of acamprosate. Exploration of the GlyR subtypes involved and efforts to develop subtype specific agonists or antagonists may offer new pharmacotherapies for alcohol use disorders.
Collapse
Affiliation(s)
- Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Helga H Lidö
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
52
|
Low dose of alcohol attenuates pro-atherosclerotic activity of thrombin. Atherosclerosis 2017; 265:215-224. [DOI: 10.1016/j.atherosclerosis.2017.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/29/2017] [Accepted: 09/01/2017] [Indexed: 01/11/2023]
|
53
|
Ethanol Stimulates Locomotion via a G αs-Signaling Pathway in IL2 Neurons in Caenorhabditis elegans. Genetics 2017; 207:1023-1039. [PMID: 28951527 PMCID: PMC5676223 DOI: 10.1534/genetics.117.300119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 09/23/2017] [Indexed: 01/21/2023] Open
Abstract
Alcohol abuse is among the top causes of preventable death, generating considerable financial, health, and societal burdens. Paradoxically, alcohol... Alcohol is a potent pharmacological agent when consumed acutely at sufficient quantities and repeated overuse can lead to addiction and deleterious effects on health. Alcohol is thought to modulate neuronal function through low-affinity interactions with proteins, in particular with membrane channels and receptors. Paradoxically, alcohol acts as both a stimulant and a sedative. The exact molecular mechanisms for the acute effects of ethanol on neurons, as either a stimulant or a sedative, however remain unclear. We investigated the role that the heat shock transcription factor HSF-1 played in determining a stimulatory phenotype of Caenorhabditis elegans in response to physiologically relevant concentrations of ethanol (17 mM; 0.1% v/v). Using genetic techniques, we demonstrate that either RNA interference of hsf-1 or use of an hsf-1(sy441) mutant lacked the enhancement of locomotion in response to acute ethanol exposure evident in wild-type animals. We identify that the requirement for HSF-1 in this phenotype was IL2 neuron-specific and required the downstream expression of the α-crystallin ortholog HSP-16.48. Using a combination of pharmacology, optogenetics, and phenotypic analyses we determine that ethanol activates a Gαs-cAMP-protein kinase A signaling pathway in IL2 neurons to stimulate nematode locomotion. We further implicate the phosphorylation of a specific serine residue (Ser322) on the synaptic protein UNC-18 as an end point for the Gαs-dependent signaling pathway. These findings establish and characterize a distinct neurosensory cell signaling pathway that determines the stimulatory action of ethanol and identifies HSP-16.48 and HSF-1 as novel regulators of this pathway.
Collapse
|
54
|
Antagonising TLR4-TRIF signalling before or after a low-dose alcohol binge during adolescence prevents alcohol drinking but not seeking behaviour in adulthood. Neuropharmacology 2017; 128:460-473. [PMID: 28947376 DOI: 10.1016/j.neuropharm.2017.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/18/2017] [Accepted: 09/19/2017] [Indexed: 12/20/2022]
Abstract
Adolescents frequently engage in risky behaviours such as binge drinking. Binge drinking, in turn, perturbs neurodevelopment reinforcing reward seeking behaviour in adulthood. Current animal models are limited in their portrayal of this behaviour and the assessment of neuroimmune involvement (specifically the role of Toll-like receptor 4 (TLR4)). Therefore, the aims of this project were to develop a more relevant animal model of adolescent alcohol exposure and to characterise its effects on TLR4 signalling and alcohol-related behaviours later life. Balb/c mice received a short (P22-P25), low dose alcohol binge during in early adolescence, and underwent tests to investigate anxiety (elevated plus maze), alcohol seeking (conditioned place preference) and binge drinking behaviour (drinking in the dark) in adulthood. Four doses of alcohol during adolescence increased alcohol-induced conditioned place preference and alcohol intake in adulthood. However, this model did not affect basal elevated plus maze performance. Subsequent analysis of nucleus accumbal mRNA, revealed increased expression of TLR4-related mRNAs in mice who received alcohol during adolescence. To further elucidate the role of TLR4, (+)-Naltrexone, a biased TLR4 antagonist was administered 30 min before or after the adolescent binge paradigm. When tested in adulthood, (+)-Naltrexone treated mice exhibited reduced alcohol intake however, alcohol seeking and anxiety behaviour was unaltered. This study highlights that even a small amount of alcohol, when given during a critical neurodevelopmental period, can potentiate alcohol-related behaviours and TLR4 activation later in life. Interestingly, attenuation of TLR4 before or after adolescent alcohol exposure reduced only binge alcohol intake in adulthood.
Collapse
|
55
|
Dual activation of neuronal G protein-gated inwardly rectifying potassium (GIRK) channels by cholesterol and alcohol. Sci Rep 2017; 7:4592. [PMID: 28676630 PMCID: PMC5496853 DOI: 10.1038/s41598-017-04681-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
Activation of G protein-gated inwardly rectifying potassium (GIRK) channels leads to a hyperpolarization of the neuron’s membrane potential, providing an important component of inhibition in the brain. In addition to the canonical G protein-activation pathway, GIRK channels are activated by small molecules but less is known about the underlying gating mechanisms. One drawback to previous studies has been the inability to control intrinsic and extrinsic factors. Here we used a reconstitution strategy with highly purified mammalian GIRK2 channels incorporated into liposomes and demonstrate that cholesterol or intoxicating concentrations of ethanol, i.e., >20 mM, each activate GIRK2 channels directly, in the absence of G proteins. Notably, both activators require the membrane phospholipid PIP2 but appear to interact independently with different regions of the channel. Elucidating the mechanisms underlying G protein-independent pathways of activating GIRK channels provides a unique strategy for developing new types of neuronal excitability modulators.
Collapse
|
56
|
Kruithof AC, Watanabe S, Peeters PA, de Kam ML, Zuiker RG, Stevens J, van Gerven JM, Stockis A. Pharmacological interactions between brivaracetam and ethanol in healthy males. J Psychopharmacol 2017; 31:915-926. [PMID: 27649776 DOI: 10.1177/0269881116665326] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This double-blind, randomized, three-way crossover study explored the potential pharmacokinetic and pharmacodynamic interactions between ethanol and brivaracetam in 18 healthy males, as required for the development of CNS-active drugs. Subjects received (A) ethanol+brivaracetam, (B) ethanol placebo+brivaracetam and (C) ethanol+brivaracetam placebo. Ethanol was infused as a 5.5-hour intravenous clamp with the first 0.5-hour as loading phase to a target level of 0.6 g/L, and brivaracetam was orally administered as a single 200 mg dose. No relevant pharmacokinetic interactions were observed. Co-administration of brivaracetam and ethanol resulted in decreased saccadic peak velocity, smooth pursuit, adaptive tracking and VAS alertness, and increased body sway, saccadic reaction time and VAS score for ethanol effect compared with brivaracetam alone or ethanol alone. Additionally, the immediate word recall scores were generally lower when brivaracetam was co-administered with ethanol, whereas the delayed word test did not show clear additional effects. A post-hoc exploratory analysis for supra-additivity suggested that most pharmacodynamic effects were likely to be additive in nature, except for adaptive tracking, which appeared to be slightly supra-additive. In conclusion, brivaracetam increased ethanol effects on psychomotor function, attention and memory in healthy males. Intake of brivaracetam with alcohol is not recommended.
Collapse
Affiliation(s)
| | | | | | | | - Rob Gja Zuiker
- 1 Centre for Human Drug Research, Leiden, the Netherlands
| | - Jasper Stevens
- 1 Centre for Human Drug Research, Leiden, the Netherlands
| | | | | |
Collapse
|
57
|
DiGuiseppi D, Milorey B, Lewis G, Kubatova N, Farrell S, Schwalbe H, Schweitzer-Stenner R. Probing the Conformation-Dependent Preferential Binding of Ethanol to Cationic Glycylalanylglycine in Water/Ethanol by Vibrational and NMR Spectroscopy. J Phys Chem B 2017; 121:5744-5758. [DOI: 10.1021/acs.jpcb.7b02899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | - Nina Kubatova
- Institut
für Organische Chemie und Chemische Biologie, Johann Wolfgang Goethe-Universität, 60438 Frankfurt am Main, Germany
| | | | - Harald Schwalbe
- Institut
für Organische Chemie und Chemische Biologie, Johann Wolfgang Goethe-Universität, 60438 Frankfurt am Main, Germany
| | | |
Collapse
|
58
|
Cui C, Koob GF. Titrating Tipsy Targets: The Neurobiology of Low-Dose Alcohol. Trends Pharmacol Sci 2017; 38:556-568. [PMID: 28372826 DOI: 10.1016/j.tips.2017.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/04/2017] [Accepted: 03/06/2017] [Indexed: 11/30/2022]
Abstract
Limited attention has been given to our understanding of how the brain responds to low-dose alcohol (ethanol) and what molecular and cellular targets mediate these effects. Even at concentrations lower than 10mM (0.046 g% blood alcohol concentration, BAC), below the legal driving limit in the USA (BAC 0.08 g%), alcohol impacts brain function and behavior. Understanding what molecular and cellular targets mediate the initial effects of alcohol and subsequent neuroplasticity could provide a better understanding of vulnerability or resilience to developing alcohol use disorders. We review here what is known about the neurobiology of low-dose alcohol, provide insights into potential molecular targets, and discuss future directions and challenges in further defining targets of low-dose alcohol at the molecular, cellular, and circuitry levels.
Collapse
Affiliation(s)
- Changhai Cui
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
59
|
Cevallos AM, Herrera J, López-Villaseñor I, Hernández R. Differential Effects of Two Widely Used Solvents, DMSO and Ethanol, on the Growth and Recovery of Trypanosoma cruzi Epimastigotes in Culture. THE KOREAN JOURNAL OF PARASITOLOGY 2017; 55:81-84. [PMID: 28285511 PMCID: PMC5365264 DOI: 10.3347/kjp.2017.55.1.81] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/15/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease. Epimastigote forms of T. cruzi can be readily cultured in axenic conditions. Ethanol and dimethyl sulfoxide (DMSO) are commonly used solvents employed as vehicles for hydrophobic compounds. In order to produce a reference plot of solvent dependent growth inhibition for T. cruzi research, the growth of epimastigotes was analyzed in the presence of different concentrations of ethanol (0.1–4.0%) and DMSO (0.5–7.5%). The ability of the parasites to resume growth after removal of these solvents was also examined. As expected, both ethanol and DMSO produced a dose-dependent inhibition of cellular growth. Parasites could recover normal growth after 9 days in up to 2% ethanol or 5% DMSO. Since DMSO was better tolerated than ethanol, it is thus recommended to prefer DMSO over ethanol in the case of a similar solubility of a given compound.
Collapse
Affiliation(s)
- Ana María Cevallos
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70-228, CP 04510, Ciudad de México, México
| | - Juliana Herrera
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70-228, CP 04510, Ciudad de México, México
| | - Imelda López-Villaseñor
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70-228, CP 04510, Ciudad de México, México
| | - Roberto Hernández
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apartado Postal 70-228, CP 04510, Ciudad de México, México
| |
Collapse
|
60
|
Roberto M, Varodayan FP. Synaptic targets: Chronic alcohol actions. Neuropharmacology 2017; 122:85-99. [PMID: 28108359 DOI: 10.1016/j.neuropharm.2017.01.013] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/23/2016] [Accepted: 01/14/2017] [Indexed: 01/02/2023]
Abstract
Alcohol acts on numerous cellular and molecular targets to regulate neuronal communication within the brain. Chronic alcohol exposure and acute withdrawal generate prominent neuroadaptations at synapses, including compensatory effects on the expression, localization and function of synaptic proteins, channels and receptors. The present article reviews the literature describing the synaptic effects of chronic alcohol exposure and their relevance for synaptic transmission in the central nervous system. This review is not meant to be comprehensive, but rather to highlight the effects that have been observed most consistently and that are thought to contribute to the development of alcohol dependence and the negative aspects of withdrawal. Specifically, we will focus on the major excitatory and inhibitory neurotransmitters in the brain, glutamate and GABA, respectively, and how their neuroadaptations after chronic alcohol exposure contributes to alcohol reinforcement, dependence and withdrawal. This article is part of the Special Issue entitled "Alcoholism".
Collapse
|
61
|
Paxman J, Hunt B, Hallan D, Zarbock SR, Woodbury DJ. Drunken Membranes: Short-Chain Alcohols Alter Fusion of Liposomes to Planar Lipid Bilayers. Biophys J 2017; 112:121-132. [PMID: 28076803 PMCID: PMC5232861 DOI: 10.1016/j.bpj.2016.11.3205] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
Although the effects of ethanol on protein receptors and lipid membranes have been studied extensively, ethanol's effect on vesicles fusing to lipid bilayers is not known. To determine the effect of alcohols on fusion rates, we utilized the nystatin/ergosterol fusion assay to measure fusion of liposomes to a planar lipid bilayer (BLM). The addition of ethanol excited fusion when applied on the cis (vesicle) side, and inhibited fusion on the trans side. Other short-chain alcohols followed a similar pattern. In general, the inhibitory effect of alcohols (trans) occurs at lower doses than the excitatory (cis) effect, with a decrease of 29% in fusion rates at the legal driving limit of 0.08% (w/v) ethanol (IC50 = 0.2% v/v, 34 mM). Similar inhibitory effects were observed with methanol, propanol, and butanol, with ethanol being the most potent. Significant variability was observed with different alcohols when applied to the cis side. Ethanol and propanol enhanced fusion, butanol also enhanced fusion but was less potent, and low doses of methanol mildly inhibited fusion. The inhibition by trans addition of alcohols implies that they alter the planar membrane structure and thereby increase the activation energy required for fusion, likely through an increase in membrane fluidity. The cis data are likely a combination of the above effect and a proportionally greater lowering of the vesicle lysis tension and hydration repulsive pressure that combine to enhance fusion. Alternate hypotheses are also discussed. The inhibitory effect of ethanol on liposome-membrane fusion is large enough to provide a possible biophysical explanation of compromised neuronal behavior.
Collapse
Affiliation(s)
- Jason Paxman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Brady Hunt
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - David Hallan
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Samuel R Zarbock
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Dixon J Woodbury
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah.
| |
Collapse
|
62
|
Voltage-Sensitive Potassium Channels of the BK Type and Their Coding Genes Are Alcohol Targets in Neurons. Handb Exp Pharmacol 2017; 248:281-309. [PMID: 29204711 DOI: 10.1007/164_2017_78] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Among all members of the voltage-gated, TM6 ion channel superfamily, the proteins that constitute calcium- and voltage-gated potassium channels of large conductance (BK) and their coding genes are unique for their involvement in ethanol-induced disruption of normal physiology and behavior. Moreover, in vitro studies document that BK activity is modified by ethanol with an EC50~23 mM, which is near blood alcohol levels considered legal intoxication in most states of the USA (0.08 g/dL = 17.4 mM). Following a succinct introduction to our current understanding of BK structure and function in central neurons, with a focus on neural circuits that contribute to the neurobiology of alcohol use disorders (AUD), we review the modifications in organ physiology by alcohol exposure via BK and the different molecular elements that determine the ethanol response of BK in alcohol-naïve systems, including the role of an ethanol-recognizing site in the BK-forming slo1 protein, modulation of accessory BK subunits, and their coding genes. The participation of these and additional elements in determining the response of a system or an organism to protracted ethanol exposure is consequently analyzed, with insights obtained from invertebrate and vertebrate models. Particular emphasis is put on the role of BK and coding genes in different forms of tolerance to alcohol exposure. We finally discuss genetic results on BK obtained in invertebrate organisms and rodents in light of possible extrapolation to human AUD.
Collapse
|
63
|
Slater CA, Jackson A, Muldoon PP, Dawson A, O'Brien M, Soll LG, Abdullah R, Carroll FI, Tapper AR, Miles MF, Banks ML, Bettinger JC, Damaj IM. Nicotine Enhances the Hypnotic and Hypothermic Effects of Alcohol in the Mouse. Alcohol Clin Exp Res 2016; 40:62-72. [PMID: 26727524 DOI: 10.1111/acer.12918] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/28/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ethanol (EtOH) and nicotine abuse are 2 leading causes of preventable mortality in the world, but little is known about the pharmacological mechanisms mediating co-abuse. Few studies have examined the interaction of the acute effects of EtOH and nicotine. Here, we examine the effects of nicotine administration on the duration of EtOH-induced loss of righting reflex (LORR) and characterize the nature of their pharmacological interactions in C57BL/6J mice. METHODS We assessed the effects of EtOH and nicotine and the nature of their interaction in the LORR test using isobolographic analysis after acute injection in C57BL/6J male mice. Next, we examined the importance of receptor efficacy using nicotinic partial agonists varenicline and sazetidine. We evaluated the involvement of major nicotinic acetylcholine receptor (nAChR) subtypes using nicotinic antagonist mecamylamine and nicotinic α4- and α7-knockout mice. The selectivity of nicotine's actions on EtOH-induced LORR was examined by testing nicotine's effects on the hypnotic properties of ketamine and pentobarbital. We also assessed the development of tolerance after repeated nicotine exposure. Last, we assessed whether the effects of nicotine on EtOH-induced LORR extend to hypothermia and EtOH intake in the drinking in the dark (DID) paradigm. RESULTS We found that acute nicotine injection enhances EtOH's hypnotic effects in a synergistic manner and that receptor efficacy plays an important role in this interaction. Furthermore, tolerance developed to the enhancement of EtOH's hypnotic effects by nicotine after repeated exposure of the drug. α4* and α7 nAChRs seem to play an important role in nicotine-EtOH interaction in the LORR test. In addition, the magnitude of EtOH-induced LORR enhancement by nicotine was more pronounced in C57BL/6J than DBA/2J mice. Furthermore, acute nicotine enhanced ketamine and pentobarbital hypnotic effects in the mouse. Finally, nicotine enhanced EtOH-induced hypothermia but decreased EtOH intake in the DID test. CONCLUSIONS Our results demonstrate that nicotine synergistically enhances EtOH-induced LORR in the mouse.
Collapse
Affiliation(s)
- Cassandra A Slater
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Asti Jackson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Pretal P Muldoon
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Anton Dawson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Megan O'Brien
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Lindsey G Soll
- Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Rehab Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, North Carolina
| | - Andrew R Tapper
- Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michael F Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Jill C Bettinger
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Imad M Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
64
|
Burgos CF, Yévenes GE, Aguayo LG. Structure and Pharmacologic Modulation of Inhibitory Glycine Receptors. Mol Pharmacol 2016; 90:318-25. [PMID: 27401877 PMCID: PMC4998662 DOI: 10.1124/mol.116.105726] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/08/2016] [Indexed: 01/08/2023] Open
Abstract
Glycine receptors (GlyR) are inhibitory Cys-loop ion channels that contribute to the control of excitability along the central nervous system (CNS). GlyR are found in the spinal cord and brain stem, and more recently they were reported in higher regions of the CNS such as the hippocampus and nucleus accumbens. GlyR are involved in motor coordination, respiratory rhythms, pain transmission, and sensory processing, and they are targets for relevant physiologic and pharmacologic modulators. Several studies with protein crystallography and cryoelectron microscopy have shed light on the residues and mechanisms associated with the activation, blockade, and regulation of pentameric Cys-loop ion channels at the atomic level. Initial studies conducted on the extracellular domain of acetylcholine receptors, ion channels from prokaryote homologs-Erwinia chrysanthemi ligand-gated ion channel (ELIC), Gloeobacter violaceus ligand-gated ion channel (GLIC)-and crystallized eukaryotic receptors made it possible to define the overall structure and topology of the Cys-loop receptors. For example, the determination of pentameric GlyR structures bound to glycine and strychnine have contributed to visualizing the structural changes implicated in the transition between the open and closed states of the Cys-loop receptors. In this review, we summarize how the new information obtained in functional, mutagenesis, and structural studies have contributed to a better understanding of the function and regulation of GlyR.
Collapse
Affiliation(s)
- Carlos F Burgos
- Laboratory of Neurophysiology (C.F.B., L.G.A.), and Laboratory of Neuropharmacology (G.E.Y.), Department of Physiology, University of Concepción, Concepción, Chile
| | - Gonzalo E Yévenes
- Laboratory of Neurophysiology (C.F.B., L.G.A.), and Laboratory of Neuropharmacology (G.E.Y.), Department of Physiology, University of Concepción, Concepción, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology (C.F.B., L.G.A.), and Laboratory of Neuropharmacology (G.E.Y.), Department of Physiology, University of Concepción, Concepción, Chile
| |
Collapse
|
65
|
San Martin L, Cerda F, Jin C, Jimenez V, Yevenes GE, Hernandez T, Nova D, Fuentealba J, Aguayo LG, Guzman L. Reversal of Ethanol-induced Intoxication by a Novel Modulator of Gβγ Protein Potentiation of the Glycine Receptor. J Biol Chem 2016; 291:18791-8. [PMID: 27402845 DOI: 10.1074/jbc.m116.740555] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 01/31/2023] Open
Abstract
The acute intoxicating effects of ethanol in the central nervous system result from the modulation of several molecular targets. It is widely accepted that ethanol enhances the activity of the glycine receptor (GlyR), thus enhancing inhibitory neurotransmission, leading to motor effects, sedation, and respiratory depression. We previously reported that small peptides interfered with the binding of Gβγ to the GlyR and consequently inhibited the ethanol-induced potentiation of the receptor. Now, using virtual screening, we identified a subset of small molecules capable of interacting with the binding site of Gβγ. One of these compounds, M554, inhibited the ethanol potentiation of the GlyR in both evoked currents and synaptic transmission in vitro When this compound was tested in vivo in mice treated with ethanol (1-3.5 g/kg), it was found to induce a faster recovery of motor incoordination in rotarod experiments and a shorter sedative effect in loss of righting reflex assays. This study describes a novel molecule that might be relevant for the design of useful therapeutic compounds in the treatment of acute alcohol intoxication.
Collapse
Affiliation(s)
| | - Fabian Cerda
- From the Laboratories of Molecular Neurobiology and
| | - Chunyang Jin
- the Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina 27709, and
| | - Veronica Jimenez
- the Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, 403901 Concepción, Chile
| | | | | | - Daniela Nova
- From the Laboratories of Molecular Neurobiology and
| | | | - Luis G Aguayo
- Neurophysiology, Department of Physiology, University of Concepción, 403901 Concepción, Chile
| | | |
Collapse
|
66
|
Zhang D, Xiong W, Jackson MF, Parkinson FE. Ethanol Tolerance Affects Endogenous Adenosine Signaling in Mouse Hippocampus. J Pharmacol Exp Ther 2016; 358:31-8. [PMID: 27189965 PMCID: PMC4931878 DOI: 10.1124/jpet.116.232231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/18/2016] [Indexed: 12/29/2022] Open
Abstract
Ethanol has many pharmacological effects, including increases in endogenous adenosine levels and adenosine receptor activity in brain. Ethanol consumption is associated with both positive and negative health outcomes, but tolerance to the behavioral effects of ethanol can lead to increased consumption, which increases the risk of negative health outcomes. The present study was performed to test whether a 7-day treatment with ethanol is linked to reduced adenosine signaling and whether this is a consequence of reduced ecto-5'-nucleotidase activity. Wild-type (CD73(+/+)) and ecto-5'-nucleotidase-deficient (CD73(-/-)) mice were treated with ethanol (2 g/kg) or saline for 7 days. In CD73(+/+) mice, repeated ethanol treatment reduced the hypothermic and ataxic effects of acute ethanol, indicating the development of tolerance to the acute effects of ethanol. In CD73(+/+) mice, this 7-day ethanol treatment led to increased hippocampal synaptic activity and reduced adenosine A1 receptor activity under both basal and low Mg(2+) conditions. These effects of ethanol tolerance were associated with an 18% decrease in activity of ecto-5'-nucleotidase activity in hippocampal cell membranes. In contrast, ethanol treatment was not associated with changes in synaptic activity or adenosine signaling in hippocampus from CD73(-/-) mice. These data indicate that ethanol treatment is associated with a reduction in adenosine signaling through adenosine A1 receptors in hippocampus, mediated, at least in part, via reduced ecto-5'-nucleotidase activity.
Collapse
Affiliation(s)
- Dali Zhang
- Department of Pharmacology and Therapeutics, University of Manitoba, and Neuroscience Research Program, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Wei Xiong
- Department of Pharmacology and Therapeutics, University of Manitoba, and Neuroscience Research Program, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Michael F Jackson
- Department of Pharmacology and Therapeutics, University of Manitoba, and Neuroscience Research Program, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Fiona E Parkinson
- Department of Pharmacology and Therapeutics, University of Manitoba, and Neuroscience Research Program, Health Sciences Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
67
|
McCracken ML, Gorini G, McCracken LM, Mayfield RD, Harris RA, Trudell JR. Inter- and Intra-Subunit Butanol/Isoflurane Sites of Action in the Human Glycine Receptor. Front Mol Neurosci 2016; 9:45. [PMID: 27378846 PMCID: PMC4906044 DOI: 10.3389/fnmol.2016.00045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/26/2016] [Indexed: 11/24/2022] Open
Abstract
Glycine receptors (GlyRs) mediate inhibitory neurotransmission and are targets for alcohols and anesthetics in brain. GlyR transmembrane (TM) domains contain critical residues for alcohol/anesthetic action: amino acid A288 in TM3 forms crosslinks with TM1 (I229) in the adjacent subunit as well as TM2 (S267) and TM4 (Y406, W407, I409, Y410) in the same subunit. We hypothesized that these residues may participate in intra-subunit and inter-subunit sites of alcohol/anesthetic action. The following double and triple mutants of GLRA1 cDNA (encoding human glycine receptor alpha 1 subunit) were injected into Xenopus laevis oocytes: I229C/A288C, I229C/A288C/C290S, A288C/Y406C, A288C/W407C, A288C/I409C, and A288C/Y410C along with the corresponding single mutants and wild-type GLRA1. Butanol (22 mM) or isoflurane (0.6 mM) potentiation of GlyR-mediated currents before and after application of the cysteine crosslinking agent HgCl2 (10 μM) was measured using two-electrode voltage clamp electrophysiology. Crosslinking nearly abolished butanol and isoflurane potentiation in the I229C/A288C and I229C/A288C/C290S mutants but had no effect in single mutants or wild-type. Crosslinking also inhibited butanol and isoflurane potentiation in the TM3-4 mutants (A288C/Y406C, A288C/W407C, A288C/I409C, A288C/Y410C) with no effect in single mutants or wild-type. We extracted proteins from oocytes expressing I229C/288C, A288C/Y410C, or wild-type GlyRs, used mass spectrometry to verify their expression and possible inter-subunit dimerization, plus immunoblotting to investigate the biochemical features of proposed crosslinks. Wild-type GlyR subunits measured about 50 kDa; after crosslinking, the dimeric/monomeric 100:50 kDa band ratio was significantly increased in I229C/288C but not A288C/Y410C mutants or wild-type, providing support for TM1-3 inter-subunit and TM3-4 intra-subunit crosslinking. A GlyR homology model based on the GluCl template provides further evidence for a multi-site model for alcohol/anesthetic interaction with human GLRA1.
Collapse
Affiliation(s)
- Mandy L McCracken
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at AustinAustin, TX, USA; Integrative Neuroscience Research Branch, Neurobiology of Addiction Section, National Institute on Drug Abuse, National Institutes of HealthBaltimore, MD, USA
| | - Giorgio Gorini
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin Austin, TX, USA
| | - Lindsay M McCracken
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin Austin, TX, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin Austin, TX, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin Austin, TX, USA
| | - James R Trudell
- Department of Anesthesia and Beckman Program for Molecular and Genetic Medicine, Stanford School of Medicine Stanford, CA, USA
| |
Collapse
|
68
|
Richards JR, Laurin EG, Bretz SW, Traylor BR, Panacek EA. Treatment of ethanol poisoning and associated hypoventilation with doxapram. Am J Emerg Med 2016; 34:2253.e1-2253.e2. [PMID: 27233697 DOI: 10.1016/j.ajem.2016.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 11/17/2022] Open
Affiliation(s)
- John R Richards
- Department of Emergency Medicine, University of California Davis Medical Center, Sacramento, CA.
| | - Erik G Laurin
- Department of Emergency Medicine, University of California Davis Medical Center, Sacramento, CA
| | | | - Brittany R Traylor
- Department of Pharmacy, University of California Davis Medical Center, Sacramento, CA
| | - Edward A Panacek
- Department of Emergency Medicine, University of South Alabama College of Medicine, Mobile, AL
| |
Collapse
|
69
|
Dopico AM, Bukiya AN, Kuntamallappanavar G, Liu J. Modulation of BK Channels by Ethanol. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:239-79. [PMID: 27238266 PMCID: PMC5257281 DOI: 10.1016/bs.irn.2016.03.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In alcohol-naïve systems, ethanol (<100mM) exposure of calcium-gated BK channels perturbs physiology and behavior. Brief (several minutes) ethanol exposure usually leads to increased BK current, which results from ethanol interaction with a pocket mapped to the BK channel-forming slo1 protein cytosolic tail domain. The importance of this region in ethanol-induced intoxication has been independently supported by an unbiased screen of Caenorhabditis elegans slo1 mutants. However, ethanol-induced BK activation is not universal as refractoriness and inhibition have been reported. The final effect depends on many factors, including intracellular calcium levels, slo1 isoform, BK beta subunit composition, posttranslational modification of BK proteins, channel lipid microenvironment, and type of ethanol administration. Studies in Drosophila melanogaster, C. elegans, and rodents show that protracted/repeated ethanol administration leads to tolerance to ethanol-induced modification of BK-driven physiology and behavior. Unveiling the mechanisms underlying tolerance is of major importance, as tolerance to ethanol has been proposed as predictor of risk for alcoholism.
Collapse
Affiliation(s)
- A M Dopico
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - A N Bukiya
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - G Kuntamallappanavar
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - J Liu
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
70
|
Davis SJ, Scott LL, Ordemann G, Philpo A, Cohn J, Pierce-Shimomura JT. Putative calcium-binding domains of the Caenorhabditis elegans BK channel are dispensable for intoxication and ethanol activation. GENES BRAIN AND BEHAVIOR 2016; 14:454-65. [PMID: 26113050 DOI: 10.1111/gbb.12229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 06/16/2015] [Accepted: 06/22/2015] [Indexed: 01/17/2023]
Abstract
Alcohol modulates the highly conserved, voltage- and calcium-activated potassium (BK) channel, which contributes to alcohol-mediated behaviors in species from worms to humans. Previous studies have shown that the calcium-sensitive domains, RCK1 and the Ca(2+) bowl, are required for ethanol activation of the mammalian BK channel in vitro. In the nematode Caenorhabditis elegans, ethanol activates the BK channel in vivo, and deletion of the worm BK channel, SLO-1, confers strong resistance to intoxication. To determine if the conserved RCK1 and calcium bowl domains were also critical for intoxication and basal BK channel-dependent behaviors in C. elegans, we generated transgenic worms that express mutated SLO-1 channels predicted to have the RCK1, Ca(2+) bowl or both domains rendered insensitive to calcium. As expected, mutating these domains inhibited basal function of SLO-1 in vivo as neck and body curvature of these mutants mimicked that of the BK null mutant. Unexpectedly, however, mutating these domains singly or together in SLO-1 had no effect on intoxication in C. elegans. Consistent with these behavioral results, we found that ethanol activated the SLO-1 channel in vitro with or without these domains. By contrast, in agreement with previous in vitro findings, C. elegans harboring a human BK channel with mutated calcium-sensing domains displayed resistance to intoxication. Thus, for the worm SLO-1 channel, the putative calcium-sensitive domains are critical for basal in vivo function but unnecessary for in vivo ethanol action.
Collapse
Affiliation(s)
- S J Davis
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - L L Scott
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - G Ordemann
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - A Philpo
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - J Cohn
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - J T Pierce-Shimomura
- Waggoner Center for Alcohol and Addiction Research, Institute for Neuroscience, Center for Learning and Memory, Center for Brain, Behavior and Evolution, and Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
71
|
Abstract
Most people who are regular consumers of psychoactive drugs are not drug addicts, nor will they ever become addicts. In neurobiological theories, non-addictive drug consumption is acknowledged only as a "necessary" prerequisite for addiction, but not as a stable and widespread behavior in its own right. This target article proposes a new neurobiological framework theory for non-addictive psychoactive drug consumption, introducing the concept of "drug instrumentalization." Psychoactive drugs are consumed for their effects on mental states. Humans are able to learn that mental states can be changed on purpose by drugs, in order to facilitate other, non-drug-related behaviors. We discuss specific "instrumentalization goals" and outline neurobiological mechanisms of how major classes of psychoactive drugs change mental states and serve non-drug-related behaviors. We argue that drug instrumentalization behavior may provide a functional adaptation to modern environments based on a historical selection for learning mechanisms that allow the dynamic modification of consummatory behavior. It is assumed that in order to effectively instrumentalize psychoactive drugs, the establishment of and retrieval from a drug memory is required. Here, we propose a new classification of different drug memory subtypes and discuss how they interact during drug instrumentalization learning and retrieval. Understanding the everyday utility and the learning mechanisms of non-addictive psychotropic drug use may help to prevent abuse and the transition to drug addiction in the future.
Collapse
|
72
|
Camarini R, Pautassi RM. Behavioral sensitization to ethanol: Neural basis and factors that influence its acquisition and expression. Brain Res Bull 2016; 125:53-78. [PMID: 27093941 DOI: 10.1016/j.brainresbull.2016.04.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/29/2022]
Abstract
Ethanol-induced behavioral sensitization (EBS) was first described in 1980, approximately 10 years after the phenomenon was described for psychostimulants. Ethanol acts on γ-aminobutyric acid (GABA) and glutamate receptors as an allosteric agonist and antagonist, respectively, but it also affects many other molecular targets. The multiplicity of factors involved in the behavioral and neurochemical effects of ethanol and the ensuing complexity may explain much of the apparent disparate results, found across different labs, regarding ethanol-induced behavioral sensitization. Although the mesocorticolimbic dopamine system plays an important role in EBS, we provide evidence of the involvement of other neurotransmitter systems, mainly the glutamatergic, GABAergic, and opioidergic systems. This review also analyses the neural underpinnings (e.g., induction of cellular transcription factors such as cyclic adenosine monophosphate response element binding protein and growth factors, such as the brain-derived neurotrophic factor) and other factors that influence the phenomenon, including age, sex, dose, and protocols of drug administration. One of the reasons that make EBS an attractive phenomenon is the assumption, firmly based on empirical evidence, that EBS and addiction-related processes have common molecular and neural basis. Therefore, EBS has been used as a model of addiction processes. We discuss the association between different measures of ethanol-induced reward and EBS. Parallels between the pharmacological basis of EBS and acute motor effects of ethanol are also discussed.
Collapse
Affiliation(s)
- Rosana Camarini
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil.
| | - Ricardo Marcos Pautassi
- Instituto de Investigaciones Médicas M. y M. Ferreyra, Córdoba (IMMF-CONICET-Universidad Nacional de Córdoba), Universidad Nacional de Córdoba, Argentina
| |
Collapse
|
73
|
Kiecker C. The chick embryo as a model for the effects of prenatal exposure to alcohol on craniofacial development. Dev Biol 2016; 415:314-325. [PMID: 26777098 DOI: 10.1016/j.ydbio.2016.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/28/2015] [Accepted: 01/13/2016] [Indexed: 12/15/2022]
Abstract
Prenatal exposure to ethanol results in fetal alcohol spectrum disorder (FASD), a syndrome characterised by a broad range of clinical manifestations including craniofacial dysmorphologies and neurological defects. The characterisation of the mechanisms by which ethanol exerts its teratogenic effects is difficult due to the pleiotropic nature of its actions. Different experimental model systems have been employed to investigate the aetiology of FASD. Here, I will review studies using these different model organisms that have helped to elucidate how ethanol causes the craniofacial abnormalities characteristic of FASD. In these studies, ethanol was found to impair the prechordal plate-an important embryonic signalling centre-during gastrulation and to negatively affect the induction, migration and survival of the neural crest, a cell population that generates the cartilage and most of the bones of the skull. At the cellular level, ethanol appears to inhibit Sonic hedgehog signalling, alter levels of retionoic acid activity, trigger a Ca(2+)-CamKII-dependent pathway that antagonises WNT signalling, affect cytoskeletal dynamics and increase oxidative stress. Embryos of the domestic chick Gallus gallus domesticus have played a central role in developing a working model for the effects of ethanol on craniofacial development because they are easily accessible and because key steps in craniofacial development are particularly well established in the avian embryo. I will finish this review by highlighting some potential future avenues of fetal alcohol research.
Collapse
Affiliation(s)
- Clemens Kiecker
- MRC Centre for Developmental Neurobiology, 4th Floor, Hodgkin Building, Guy's Hospital Campus, King's College London, UK.
| |
Collapse
|
74
|
Follesa P, Floris G, Asuni GP, Ibba A, Tocco MG, Zicca L, Mercante B, Deriu F, Gorini G. Chronic Intermittent Ethanol Regulates Hippocampal GABA(A) Receptor Delta Subunit Gene Expression. Front Cell Neurosci 2015; 9:445. [PMID: 26617492 PMCID: PMC4637418 DOI: 10.3389/fncel.2015.00445] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/26/2015] [Indexed: 12/26/2022] Open
Abstract
Chronic ethanol consumption causes structural and functional reorganization in the hippocampus and induces alterations in the gene expression of gamma-aminobutyric acid type A receptors (GABAARs). Distinct forced intermittent exposure models have been used previously to investigate changes in GABAAR expression, with contrasting results. Here, we used repeated cycles of a Chronic Intermittent Ethanol paradigm to examine the relationship between voluntary, dependence-associated ethanol consumption, and GABAAR gene expression in mouse hippocampus. Adult male C57BL/6J mice were exposed to four 16-h ethanol vapor (or air) cycles in inhalation chambers alternated with limited-access two-bottle choice between ethanol (15%) and water consumption. The mice exposed to ethanol vapor showed significant increases in ethanol consumption compared to their air-matched controls. GABAAR alpha4 and delta subunit gene expression were measured by qRT-PCR at different stages. There were significant changes in GABAAR delta subunit transcript levels at different time points in ethanol-vapor exposed mice, while the alpha4 subunit levels remained unchanged. Correlated concurrent blood ethanol concentrations suggested that GABAAR delta subunit mRNA levels fluctuate depending on ethanol intoxication, dependence, and withdrawal state. Using a vapor-based Chronic Intermittent Ethanol procedure with combined two-bottle choice consumption, we corroborated previous evidences showing that discontinuous ethanol exposure affects GABAAR delta subunit expression but we did not observe changes in alpha4 subunit. These findings indicate that hippocampal GABAAR delta subunit expression changes transiently over the course of a Chronic Intermittent Ethanol paradigm associated with voluntary intake, in response to ethanol-mediated disturbance of GABAergic neurotransmission.
Collapse
Affiliation(s)
- Paolo Follesa
- Department of Life and Environmental Sciences, University of Cagliari Cagliari, Italy
| | - Gabriele Floris
- Department of Life and Environmental Sciences, University of Cagliari Cagliari, Italy
| | - Gino P Asuni
- Department of Life and Environmental Sciences, University of Cagliari Cagliari, Italy
| | - Antonio Ibba
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari Cagliari, Italy
| | - Maria G Tocco
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari Cagliari, Italy
| | - Luca Zicca
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari Cagliari, Italy
| | | | - Franca Deriu
- Department of Biomedical Sciences, University of Sassari Sassari, Italy
| | - Giorgio Gorini
- Department of Life and Environmental Sciences, University of Cagliari Cagliari, Italy
| |
Collapse
|
75
|
Transcriptome organization for chronic alcohol abuse in human brain. Mol Psychiatry 2015; 20:1438-47. [PMID: 25450227 PMCID: PMC4452464 DOI: 10.1038/mp.2014.159] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 12/18/2022]
Abstract
Alcohol dependence is a heterogeneous psychiatric disorder characterized by high genetic heritability and neuroadaptations occurring from repeated drug exposure. Through an integrated systems approach we observed consistent differences in transcriptome organization within postmortem human brain tissue associated with the lifetime consumption of alcohol. Molecular networks, determined using high-throughput RNA sequencing, for drinking behavior were dominated by neurophysiological targets and signaling mechanisms of alcohol. The systematic structure of gene sets demonstrates a novel alliance of multiple ion channels, and related processes, underlying lifetime alcohol consumption. Coordinate expression of these transcripts was enriched for genome-wide association signals in alcohol dependence and a meta-analysis of alcohol self-administration in mice. Further dissection of genes within alcohol consumption networks revealed the potential interaction of alternatively spliced transcripts. For example, expression of a human-specific isoform of the voltage-gated sodium channel subunit SCN4B was significantly correlated to lifetime alcohol consumption. Overall, our work demonstrates novel convergent evidence for biological networks related to excessive alcohol consumption, which may prove fundamentally important in the development of pharmacotherapies for alcohol dependence.
Collapse
|
76
|
Hillmer AT, Mason GF, Fucito LM, O'Malley SS, Cosgrove KP. How Imaging Glutamate, γ-Aminobutyric Acid, and Dopamine Can Inform the Clinical Treatment of Alcohol Dependence and Withdrawal. Alcohol Clin Exp Res 2015; 39:2268-82. [PMID: 26510169 DOI: 10.1111/acer.12893] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/29/2015] [Indexed: 12/29/2022]
Abstract
Neuroimaging studies have dramatically advanced our understanding of the neurochemical basis of alcohol dependence, a major public health issue. In this paper, we review the research generated from neurochemical specific imaging modalities including magnetic resonance spectroscopy, positron emission tomography, and single-photon emission computed tomography in studies of alcohol dependence and withdrawal. We focus on studies interrogating γ-aminobutyric acid (GABA), glutamate, and dopamine, as these are prominent neurotransmitter systems implicated in alcohol dependence. Highlighted findings include diminished dopaminergic functioning and modulation of the GABA system by tobacco smoking during alcohol withdrawal. Then, we consider how these findings impact the clinical treatment of alcohol dependence and discuss directions for future experiments to address existing gaps in the literature, for example, sex differences and smoking comorbidity. These and other considerations provide opportunities to build upon the current neurochemistry imaging literature of alcohol dependence and withdrawal, which may usher in improved therapeutic and relapse prevention strategies.
Collapse
Affiliation(s)
- Ansel T Hillmer
- Departments of Psychiatry and Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Graeme F Mason
- Departments of Psychiatry and Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Lisa M Fucito
- Departments of Psychiatry and Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Stephanie S O'Malley
- Departments of Psychiatry and Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly P Cosgrove
- Departments of Psychiatry and Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
77
|
Huang H, He Z, Zhu C, Liu L, Kou H, Shen L, Wang H. Prenatal ethanol exposure-induced adrenal developmental abnormality of male offspring rats and its possible intrauterine programming mechanisms. Toxicol Appl Pharmacol 2015; 288:84-94. [DOI: 10.1016/j.taap.2015.07.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 02/03/2023]
|
78
|
Müller CA, Geisel O, Pelz P, Higl V, Krüger J, Stickel A, Beck A, Wernecke KD, Hellweg R, Heinz A. High-dose baclofen for the treatment of alcohol dependence (BACLAD study): a randomized, placebo-controlled trial. Eur Neuropsychopharmacol 2015; 25:1167-77. [PMID: 26048580 DOI: 10.1016/j.euroneuro.2015.04.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 03/13/2015] [Accepted: 04/01/2015] [Indexed: 01/17/2023]
Abstract
Previous randomized, placebo-controlled trials (RCTs) assessing the efficacy of the selective γ-aminobutyric acid (GABA)-B receptor agonist baclofen in the treatment of alcohol dependence have reported divergent results, possibly related to the low to medium dosages of baclofen used in these studies (30-80mg/d). Based on preclinical observations of a dose-dependent effect and positive case reports in alcohol-dependent patients, the present RCT aimed to assess the efficacy and safety of individually titrated high-dose baclofen for the treatment of alcohol dependence. Out of 93 alcohol-dependent patients initially screened, 56 were randomly assigned to a double-blind treatment with individually titrated baclofen or placebo using dosages of 30-270mg/d. The multiple primary outcome measures were (1) total abstinence and (2) cumulative abstinence duration during a 12-week high-dose phase. More patients of the baclofen group maintained total abstinence during the high-dose phase than those receiving placebo (15/22, 68.2% vs. 5/21, 23.8%, p=0.014). Cumulative abstinence duration was significantly higher in patients given baclofen compared to patients of the placebo group (mean 67.8 (SD 30) vs. 51.8 (SD 29.6) days, p=0.047). No drug-related serious adverse events were observed during the trial. Individually titrated high-dose baclofen effectively supported alcohol-dependent patients in maintaining alcohol abstinence and showed a high tolerability, even in the event of relapse. These results provide further evidence for the potential of baclofen, thereby possibly extending the current pharmacological treatment options in alcohol dependence.
Collapse
Affiliation(s)
- Christian A Müller
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Olga Geisel
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Patricia Pelz
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Verena Higl
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Josephine Krüger
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Anna Stickel
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Anne Beck
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | - Rainer Hellweg
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
79
|
Abstract
BACKGROUND Alcohol regulates the expression and function of protein kinase C epsilon (PKCε). In a previous study we identified an alcohol binding site in the C1B, one of the twin C1 subdomains of PKCε (Das et al., Biochem. J., 421, 405-13, 2009). METHODS In this study, we investigated alcohol binding in the entire C1 domain (combined C1A and C1B) of PKCε. Fluorescent phorbol ester, SAPD and fluorescent diacylglycerol (DAG) analog, dansyl-DAG were used to study the effect of ethanol, butanol, and octanol on the ligand binding using fluorescence resonance energy transfer (FRET). To identify alcohol binding site(s), PKCεC1 was photolabeled with 3-azibutanol and 3-azioctanol, and analyzed by mass spectrometry. The effects of alcohols and the azialcohols on PKCε were studied in NG108-15 cells. RESULTS In the presence of alcohol, SAPD and dansyl-DAG showed different extent of FRET, indicating differential effects of alcohol on the C1A and C1B subdomains. Effects of alcohols and azialcohols on PKCε in NG108-15 cells were comparable. Azialcohols labeled Tyr-176 of C1A and Tyr-250 of C1B. Inspection of the model structure of PKCεC1 reveals that these residues are 40Å apart from each other indicating that these residues form two different alcohol binding sites. CONCLUSIONS The present results provide evidence for the presence of multiple alcohol-binding sites on PKCε and underscore the importance of targeting this PKC isoform in developing alcohol antagonists.
Collapse
Affiliation(s)
- Satyabrata Pany
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States.
| |
Collapse
|
80
|
Burgos CF, Muñoz B, Guzman L, Aguayo LG. Ethanol effects on glycinergic transmission: From molecular pharmacology to behavior responses. Pharmacol Res 2015; 101:18-29. [PMID: 26158502 DOI: 10.1016/j.phrs.2015.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
It is well accepted that ethanol is able to produce major health and economic problems associated to its abuse. Because of its intoxicating and addictive properties, it is necessary to analyze its effect in the central nervous system. However, we are only now learning about the mechanisms controlling the modification of important membrane proteins such as ligand-activated ion channels by ethanol. Furthermore, only recently are these effects being correlated to behavioral changes. Current studies show that the glycine receptor (GlyR) is a susceptible target for low concentrations of ethanol (5-40mM). GlyRs are relevant for the effects of ethanol because they are found in the spinal cord and brain stem where they primarily express the α1 subunit. More recently, the presence of GlyRs was described in higher regions, such as the hippocampus and nucleus accumbens, with a prevalence of α2/α3 subunits. Here, we review data on the following aspects of ethanol effects on GlyRs: (1) direct interaction of ethanol with amino acids in the extracellular or transmembrane domains, and indirect mechanisms through the activation of signal transduction pathways; (2) analysis of α2 and α3 subunits having different sensitivities to ethanol which allows the identification of structural requirements for ethanol modulation present in the intracellular domain and C-terminal region; (3) Genetically modified knock-in mice for α1 GlyRs that have an impaired interaction with G protein and demonstrate reduced ethanol sensitivity without changes in glycinergic transmission; and (4) GlyRs as potential therapeutic targets.
Collapse
Affiliation(s)
- Carlos F Burgos
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, Chile
| | - Braulio Muñoz
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, Chile
| | - Leonardo Guzman
- Laboratory of Molecular Neurobiology, Department of Physiology, University of Concepción, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, Chile.
| |
Collapse
|
81
|
Farris SP, Harris RA, Ponomarev I. Epigenetic modulation of brain gene networks for cocaine and alcohol abuse. Front Neurosci 2015; 9:176. [PMID: 26041984 PMCID: PMC4438259 DOI: 10.3389/fnins.2015.00176] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/30/2015] [Indexed: 12/19/2022] Open
Abstract
Cocaine and alcohol are two substances of abuse that prominently affect the central nervous system (CNS). Repeated exposure to cocaine and alcohol leads to longstanding changes in gene expression, and subsequent functional CNS plasticity, throughout multiple brain regions. Epigenetic modifications of histones are one proposed mechanism guiding these enduring changes to the transcriptome. Characterizing the large number of available biological relationships as network models can reveal unexpected biochemical relationships. Clustering analysis of variation from whole-genome sequencing of gene expression (RNA-Seq) and histone H3 lysine 4 trimethylation (H3K4me3) events (ChIP-Seq) revealed the underlying structure of the transcriptional and epigenomic landscape within hippocampal postmortem brain tissue of drug abusers and control cases. Distinct sets of interrelated networks for cocaine and alcohol abuse were determined for each abusive substance. The network approach identified subsets of functionally related genes that are regulated in agreement with H3K4me3 changes, suggesting cause and effect relationships between this epigenetic mark and gene expression. Gene expression networks consisted of recognized substrates for addiction, such as the dopamine- and cAMP-regulated neuronal phosphoprotein PPP1R1B/DARPP-32 and the vesicular glutamate transporter SLC17A7/VGLUT1 as well as potentially novel molecular targets for substance abuse. Through a systems biology based approach our results illustrate the utility of integrating epigenetic and transcript expression to establish relevant biological networks in the human brain for addiction. Future work with laboratory models may clarify the functional relevance of these gene networks for cocaine and alcohol, and provide a framework for the development of medications for the treatment of addiction.
Collapse
Affiliation(s)
- Sean P Farris
- Waggoner Center for Alcohol & Addiction Research and The College of Pharmacy, University of Texas at Austin Austin, TX, USA
| | - Robert A Harris
- Waggoner Center for Alcohol & Addiction Research and The College of Pharmacy, University of Texas at Austin Austin, TX, USA
| | - Igor Ponomarev
- Waggoner Center for Alcohol & Addiction Research and The College of Pharmacy, University of Texas at Austin Austin, TX, USA
| |
Collapse
|
82
|
June HL, Liu J, Warnock KT, Bell KA, Balan I, Bollino D, Puche A, Aurelian L. CRF-amplified neuronal TLR4/MCP-1 signaling regulates alcohol self-administration. Neuropsychopharmacology 2015; 40:1549-59. [PMID: 25567426 PMCID: PMC4397415 DOI: 10.1038/npp.2015.4] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022]
Abstract
Alcohol dependence is a complex disorder that initiates with episodes of excessive alcohol drinking known as binge drinking. It has a 50-60% risk contribution from inherited susceptibility genes; however, their exact identity and function are still poorly understood. We report that alcohol-preferring P rats have innately elevated levels of Toll-like receptor 4 (TLR4) and monocyte chemotactic protein-1 (MCP-1) that colocalize in neurons from the central nucleus of the amygdala (CeA) and ventral tegmental area (VTA). To examine the potential role of a TLR4/MCP-1 signal, we used Herpes Simplex Virus (HSV) vectors (amplicons) that retain in vivo neurotropism. Infusion of amplicons for TLR4 or MCP-1 siRNA into the CeA or VTA from the P rats inhibited target gene expression and blunted binge drinking. A similarly delivered amplicon for scrambled siRNA did not inhibit TLR4 or MCP-1 expression nor reduce binge drinking, identifying a neuronal TLR4/MCP-1 signal that regulates the initiation of voluntary alcohol self-administration. The signal was sustained during alcohol drinking by increased expression of corticotropin-releasing factor and its feedback regulation of TLR4 expression, likely contributing to the transition to alcohol dependence.
Collapse
Affiliation(s)
- Harry L June
- Neuropsychopharmacology Laboratory, Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC, USA
| | - Juan Liu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kaitlin T Warnock
- Neuropsychopharmacology Laboratory, Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC, USA
| | - Kimberly A Bell
- Neuropsychopharmacology Laboratory, Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC, USA
| | - Irina Balan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dominique Bollino
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adam Puche
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laure Aurelian
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA,Department of Pharmacology and Experimental Therapeutics, University of Maryland, 655 West Baltimore Street, Baltimore, MD 21201, USA, Tel: +1 410 7063895, Fax: +1 410 7062513, E-mail:
| |
Collapse
|
83
|
Chong Y, Kleinhammes A, Tang P, Xu Y, Wu Y. Dominant Alcohol-Protein Interaction via Hydration-Enabled Enthalpy-Driven Binding Mechanism. J Phys Chem B 2015; 119:5367-75. [PMID: 25856773 DOI: 10.1021/acs.jpcb.5b00378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Water plays an important role in weak associations of small drug molecules with proteins. Intense focus has been on binding-induced structural changes in the water network surrounding protein binding sites, especially their contributions to binding thermodynamics. However, water is also tightly coupled to protein conformations and dynamics, and so far little is known about the influence of water-protein interactions on ligand binding. Alcohols are a type of low-affinity drugs, and it remains unclear how water affects alcohol-protein interactions. Here, we present alcohol adsorption isotherms under controlled protein hydration using in situ NMR detection. As functions of hydration level, Gibbs free energy, enthalpy, and entropy of binding were determined from the temperature dependence of isotherms. Two types of alcohol binding were found. The dominant type is low-affinity nonspecific binding, which is strongly dependent on temperature and the level of hydration. At low hydration levels, this nonspecific binding only occurs above a threshold of alcohol vapor pressure. An increased hydration level reduces this threshold, with it finally disappearing at a hydration level of h ≈ 0.2 (g water/g protein), gradually shifting alcohol binding from an entropy-driven to an enthalpy-driven process. Water at charged and polar groups on the protein surface was found to be particularly important in enabling this binding. Although further increase in hydration has smaller effects on the changes of binding enthalpy and entropy, it results in a significant negative change in Gibbs free energy due to unmatched enthalpy-entropy compensation. These results show the crucial role of water-protein interplay in alcohol binding.
Collapse
Affiliation(s)
- Yuan Chong
- †Department of Physics and Astronomy, University of North Carolina, Chapel Hill, North Carolina 27599-3255, United States
| | - Alfred Kleinhammes
- †Department of Physics and Astronomy, University of North Carolina, Chapel Hill, North Carolina 27599-3255, United States
| | | | | | - Yue Wu
- †Department of Physics and Astronomy, University of North Carolina, Chapel Hill, North Carolina 27599-3255, United States
| |
Collapse
|
84
|
Sánchez A, Yévenes GE, San Martin L, Burgos CF, Moraga-Cid G, Harvey RJ, Aguayo LG. Control of ethanol sensitivity of the glycine receptor α3 subunit by transmembrane 2, the intracellular splice cassette and C-terminal domains. J Pharmacol Exp Ther 2015; 353:80-90. [PMID: 25589412 PMCID: PMC4366752 DOI: 10.1124/jpet.114.221143] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/13/2015] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that the effect of ethanol onglycine receptors (GlyRs) containing the a1 subunit is affected by interaction with heterotrimeric G proteins (Gβγ). GlyRs containing the α3 subunit are involved in inflammatory pain sensitization and rhythmic breathing and have received much recent attention. For example, it is unknown whether ethanol affects the function of this important GlyR subtype. Electrophysiologic experiments showed that GlyR α3 subunits were not potentiated by pharmacologic concentrations of ethanol or by Gβγ. Thus, we studied GlyR α1–α3 chimeras and mutants to determine the molecular properties that confer ethanol insensitivity. Mutation of corresponding glycine 254 in transmembrane domain 2 (TM2) found in a1 in the α3(A254G) –α1 chimera induced a glycine-evoked current that displayed potentiation during application of ethanol (46 ± 5%, 100 mM) and Gβγ activation (80 ± 17%). Interestingly,insertion of the intracellular α3L splice cassette into GlyR α1 abolished the enhancement of the glycine-activated current by ethanol (5 ± 6%) and activation by Gβγ (21 6 7%). In corporation of the GlyR α1 C terminus into the ethanol-resistant α3S(A254G) mutant produced a construct that displayed potentiation of the glycine-activated current with 100 mM ethanol (40 ± 6%)together with a current enhancement after G protein activation (68 ± 25%). Taken together, these data demonstrate that GlyRα3 subunits are not modulated by ethanol. Residue A254 in TM2, the α3L splice cassette, and the C-terminal domain of α3GlyRs are determinants for low ethanol sensitivity and form the molecular basis of subtype-selective modulation of GlyRs by alcohol.
Collapse
Affiliation(s)
- Andrea Sánchez
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, Concepción, Chile
| | | | | | | | | | | | | |
Collapse
|
85
|
|
86
|
O’Tousa DS, Warnock KT, Matson LM, Namjoshi OA, Halcomb ME, Cook J, Grahame NJ, June HL. Triple monoamine uptake inhibitors demonstrate a pharmacologic association between excessive drinking and impulsivity in high-alcohol-preferring (HAP) mice. Addict Biol 2015; 20:236-47. [PMID: 24118509 PMCID: PMC3984927 DOI: 10.1111/adb.12100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Approximately 30% of current drinkers in the United States drink excessively, and are referred to as problem/hazardous drinkers. These individuals, who may not meet criteria for alcohol abuse or dependence, comprise binge, heavy drinkers, or both. Given their high prevalence, interventions that reduce the risk of binge and heavy drinking have important public health implications. Impulsivity has been repeatedly associated with excessive drinking in the clinical literature. As impulsivity is correlated with, and may play a critical role in, the initiation and maintenance of excessive drinking, this behavior may be an important target for therapeutic intervention. Hence, a better understanding of pharmacological treatments capable of attenuating excessive drinking and impulsivity may markedly improve clinical outcomes. The high-alcohol-preferring (HAP) mice represent a strong rodent model to study the relationship between impulsivity and excessive alcohol drinking, as recent evidence indicates they consume high levels of alcohol throughout their active cycle and are innately impulsive. Using this model, the present study demonstrates that the triple monoamine uptake inhibitors (TUIs) amitifadine and DOV 102, 677 effectively attenuate binge drinking, heavy drinking assessed via a 24-hour free-choice assay, and impulsivity measured by the delay discounting procedure. In contrast, 3-PBC, a GABA-A α1 preferring ligand with mixed agonist-antagonist properties, attenuates excessive drinking without affecting impulsivity. These findings suggest that in HAP mice, monoamine pathways may predominate as a common mechanism underlying impulsivity and excessive drinking, while the GABAergic system may be more salient in regulating excessive drinking. We further propose that TUIs such as amitifadine and DOV 102, 677 may be used to treat the co-occurrence of impulsivity and excessive drinking.
Collapse
Affiliation(s)
- David S. O’Tousa
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN 46202
| | - Kaitlin T. Warnock
- Neuropsychopharmacology Laboratory, Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20060, USA
| | - Liana M. Matson
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN 46202
| | - Ojas A. Namjoshi
- Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Meredith E. Halcomb
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN 46202
| | - James Cook
- Department of Chemistry, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA
| | - Nicholas J. Grahame
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN 46202
| | - Harry L. June
- Neuropsychopharmacology Laboratory, Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20060, USA
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20060, USA
| |
Collapse
|
87
|
Burgos CF, Castro PA, Mariqueo T, Bunster M, Guzmán L, Aguayo LG. Evidence for α-helices in the large intracellular domain mediating modulation of the α1-glycine receptor by ethanol and Gβγ. J Pharmacol Exp Ther 2015; 352:148-55. [PMID: 25339760 PMCID: PMC4279101 DOI: 10.1124/jpet.114.217976] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 10/21/2014] [Indexed: 12/19/2022] Open
Abstract
The α1-subunit containing glycine receptors (GlyRs) is potentiated by ethanol, in part, by intracellular Gβγ actions. Previous studies have suggested that molecular requirements in the large intracellular domain are involved; however, the lack of structural data about this region has made it difficult to describe a detailed mechanism. Using circular dichroism and molecular modeling, we generated a full model of the α1-GlyR, which includes the large intracellular domain and provides new information on structural requirements for allosteric modulation by ethanol and Gβγ. The data strongly suggest the existence of an α-helical conformation in the regions near transmembrane (TM)-3 and TM4 of the large intracellular domain. The secondary structure in the N-terminal region of the large intracellular domain near TM3 appeared critical for ethanol action, and this was tested using the homologous domain of the γ2-subunit of the GABAA receptor predicted to have little helical conformation. This region of γ2 was able to bind Gβγ and form a functional channel when combined with α1-GlyR, but it was not sensitive to ethanol. Mutations in the N- and C-terminal regions introduced to replace corresponding amino acids of the α1-GlyR sequence restored the ability to be modulated by ethanol and Gβγ. Recovery of the sensitivity to ethanol was associated with the existence of a helical conformation similar to α1-GlyR, thus being an essential secondary structural requirement for GlyR modulation by ethanol and G protein.
Collapse
Affiliation(s)
- Carlos F Burgos
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| | - Patricio A Castro
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| | - Trinidad Mariqueo
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| | - Marta Bunster
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| | - Leonardo Guzmán
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology (C.F.B., .P.A.C., T.M., L.G.A.), Laboratory of Molecular Neurobiology, Department of Physiology (L.G.), Laboratory of Molecular Biophysics, Department of Biochemistry and Molecular Biology (M.B.), and Ph.D. program in Pharmacology (T.M.), University of Concepción, Concepción, Chile
| |
Collapse
|
88
|
Odeon MM, Andreu M, Yamauchi L, Grosman M, Acosta GB. Chronic postnatal stress induces voluntary alcohol intake and modifies glutamate transporters in adolescent rats. Stress 2015; 18:427-34. [PMID: 26037264 DOI: 10.3109/10253890.2015.1041909] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Postnatal stress alters stress responses for life, with serious consequences on the central nervous system (CNS), involving glutamatergic neurotransmission and development of voluntary alcohol intake. Several drugs of abuse, including alcohol and cocaine, alter glutamate transport (GluT). Here, we evaluated effects of chronic postnatal stress (CPS) on alcohol intake and brain glutamate uptake and transporters in male adolescent Wistar rats. For CPS from postnatal day (PD) 7, pups were separated from their mothers and exposed to cold stress (4 °C) for 1 h daily for 20 days; controls remained with their mothers. Then they were exposed to either voluntary ethanol (6%) or dextrose (1%) intake for 7 days (5-7 rats per group), then killed. CPS: (1) increased voluntary ethanol intake, (2) did not affect body weight gain or produce signs of toxicity with alcohol exposure, (3) increased glutamate uptake by hippocampal synaptosomes in vitro and (4) reduced protein levels (Western measurements) in hippocampus and frontal cortex of glial glutamate transporter-1 (GLT-1) and excitatory amino-acid transporter-3 (EAAT-3) but increased glutamate aspartate transporter (GLAST) levels. We propose that CPS-induced decrements in GLT-1 and EAAT-3 expression levels are opposed by activation of a compensatory mechanism to prevent excitotoxicity. A greater role for GLAST in total glutamate uptake to prevent enlarged extracellular glutamate levels is inferred. Although CPS strongly increased intake of ethanol, this had little impact on effects of CPS on brain glutamate uptake or transporters. However, the impact of early life adverse events on glutamatergic neurotransmission may underlie increased alcohol consumption in adulthood.
Collapse
Affiliation(s)
- María Mercedes Odeon
- a Institute of Pharmacological Research (ININFA), National Scientific and Technologic Research Council (CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina and
| | - Marcela Andreu
- b Laboratorio Bioquímica Médica SRL , Buenos Aires , Argentina
| | - Laura Yamauchi
- b Laboratorio Bioquímica Médica SRL , Buenos Aires , Argentina
| | | | - Gabriela Beatriz Acosta
- a Institute of Pharmacological Research (ININFA), National Scientific and Technologic Research Council (CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina and
| |
Collapse
|
89
|
Bhandage AK, Jin Z, Bazov I, Kononenko O, Bakalkin G, Korpi ER, Birnir B. GABA-A and NMDA receptor subunit mRNA expression is altered in the caudate but not the putamen of the postmortem brains of alcoholics. Front Cell Neurosci 2014; 8:415. [PMID: 25538565 PMCID: PMC4257153 DOI: 10.3389/fncel.2014.00415] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/15/2014] [Indexed: 01/16/2023] Open
Abstract
Chronic consumption of alcohol by humans has been shown to lead to impairment of executive and cognitive functions. Here, we have studied the mRNA expression of ion channel receptors for glutamate and GABA in the dorsal striatum of post-mortem brains from alcoholics (n = 29) and normal controls (n = 29), with the focus on the caudate nucleus that is associated with the frontal cortex executive functions and automatic thinking and on the putamen area that is linked to motor cortices and automatic movements. The results obtained by qPCR assay revealed significant changes in the expression of specific excitatory ionotropic glutamate and inhibitory GABA-A receptor subunit genes in the caudate but not the putamen. Thus, in the caudate we found reduced levels of mRNAs encoding the GluN2A glutamate receptor and the δ, ε, and ρ2 GABA-A receptor subunits, and increased levels of the mRNAs encoding GluD1, GluD2, and GABA-A γ1 subunits in the alcoholics as compared to controls. Interestingly in the controls, 11 glutamate and 5 GABA-A receptor genes were more prominently expressed in the caudate than the putamen (fold-increase varied from 1.24 to 2.91). Differences in gene expression patterns between the striatal regions may underlie differences in associated behavioral outputs. Our results suggest an altered balance between caudate-mediated voluntarily controlled and automatic behaviors in alcoholics, including diminished executive control on goal-directed alcohol-seeking behavior.
Collapse
Affiliation(s)
- Amol K Bhandage
- Molecular Physiology and Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| | - Zhe Jin
- Molecular Physiology and Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| | - Igor Bazov
- Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland
| | - Olga Kononenko
- Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland
| | - Georgy Bakalkin
- Pharmacology, Institute of Biomedicine, University of Helsinki Helsinki, Finland
| | - Esa R Korpi
- Department of Pharmaceutical Bioscience (Biological Research on Drug Dependence), Biomedical Center, Uppsala University Uppsala, Sweden
| | - Bryndis Birnir
- Molecular Physiology and Neuroscience, Biomedical Center, Uppsala University Uppsala, Sweden
| |
Collapse
|
90
|
Naito A, Muchhala KH, Asatryan L, Trudell JR, Homanics GE, Perkins DI, Davies DL, Alkana RL. Glycine and GABA(A) ultra-sensitive ethanol receptors as novel tools for alcohol and brain research. Mol Pharmacol 2014; 86:635-46. [PMID: 25245406 PMCID: PMC4244596 DOI: 10.1124/mol.114.093773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/19/2014] [Indexed: 11/22/2022] Open
Abstract
A critical obstacle to developing effective medications to prevent and/or treat alcohol use disorders is the lack of specific knowledge regarding the plethora of molecular targets and mechanisms underlying alcohol (ethanol) action in the brain. To identify the role of individual receptor subunits in ethanol-induced behaviors, we developed a novel class of ultra-sensitive ethanol receptors (USERs) that allow activation of a single receptor subunit population sensitized to extremely low ethanol concentrations. USERs were created by mutating as few as four residues in the extracellular loop 2 region of glycine receptors (GlyRs) or γ-aminobutyric acid type A receptors (GABA(A)Rs), which are implicated in causing many behavioral effects linked to ethanol abuse. USERs, expressed in Xenopus oocytes and tested using two-electrode voltage clamp, demonstrated an increase in ethanol sensitivity of 100-fold over wild-type receptors by significantly decreasing the threshold and increasing the magnitude of ethanol response, without altering general receptor properties including sensitivity to the neurosteroid, allopregnanolone. These profound changes in ethanol sensitivity were observed across multiple subunits of GlyRs and GABA(A)Rs. Collectively, our studies set the stage for using USER technology in genetically engineered animals as a unique tool to increase understanding of the neurobiological basis of the behavioral effects of ethanol.
Collapse
Affiliation(s)
- Anna Naito
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Karan H Muchhala
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Liana Asatryan
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - James R Trudell
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Gregg E Homanics
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Daya I Perkins
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Daryl L Davies
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| | - Ronald L Alkana
- Department of Pharmacology and Pharmaceutical Sciences (A.N., K.H.M., R.L.A.) and Titus Family Department of Clinical Pharmacy and Pharmaceutical Economics and Policy (L.A., D.L.D.), University of Southern California School of Pharmacy, Los Angeles, California; Department of Anesthesia, Beckman Program for Molecular and Genetic Medicine, Stanford University, Stanford University Medical Center, Stanford, California (J.R.T.); Departments of Anesthesiology and Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (G.E.H.); and Amgen Inc., Thousand Oaks, California (D.I.P.)
| |
Collapse
|
91
|
Fry JD. Mechanisms of naturally evolved ethanol resistance in Drosophila melanogaster. J Exp Biol 2014; 217:3996-4003. [PMID: 25392459 PMCID: PMC4229365 DOI: 10.1242/jeb.110510] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/21/2014] [Indexed: 02/02/2023]
Abstract
The decaying fruit in which Drosophila melanogaster feed and breed can contain ethanol in concentrations as high as 6-7%. In this cosmopolitan species, populations from temperate regions are consistently more resistant to ethanol poisoning than populations from the tropics, but little is known about the physiological basis of this difference. I show that when exposed to low levels of ethanol vapor, flies from a tropical African population accumulated 2-3 times more internal ethanol than flies from a European population, giving evidence that faster ethanol catabolism by European flies contributes to the resistance difference. Using lines differing only in the origin of their third chromosome, however, I show that faster ethanol elimination cannot fully explain the resistance difference, because relative to African third chromosomes, European third chromosomes confer substantially higher ethanol resistance, while having little effect on internal ethanol concentrations. European third chromosomes also confer higher resistance to acetic acid, a metabolic product of ethanol, than African third chromosomes, suggesting that the higher ethanol resistance conferred by the former might be due to increased resistance to deleterious effects of ethanol-derived acetic acid. In support of this hypothesis, when ethanol catabolism was blocked with an Alcohol dehydrogenase mutant, there was no difference in ethanol resistance between flies with European and African third chromosomes.
Collapse
Affiliation(s)
- James D Fry
- Department of Biology, University of Rochester, Rochester, NY 14627-0211, USA
| |
Collapse
|
92
|
Developmental ethanol exposure leads to dysregulation of lipid metabolism and oxidative stress in Drosophila. G3-GENES GENOMES GENETICS 2014; 5:49-59. [PMID: 25387828 PMCID: PMC4291469 DOI: 10.1534/g3.114.015040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ethanol exposure during development causes an array of developmental abnormalities, both physiological and behavioral. In mammals, these abnormalities are collectively known as fetal alcohol effects (FAE) or fetal alcohol spectrum disorder (FASD). We have established a Drosophila melanogaster model of FASD and have previously shown that developmental ethanol exposure in flies leads to reduced expression of insulin-like peptides (dILPs) and their receptor. In this work, we link that observation to dysregulation of fatty acid metabolism and lipid accumulation. Further, we show that developmental ethanol exposure in Drosophila causes oxidative stress, that this stress is a primary cause of the developmental lethality and delay associated with ethanol exposure, and, finally, that one of the mechanisms by which ethanol increases oxidative stress is through abnormal fatty acid metabolism. These data suggest a previously uncharacterized mechanism by which ethanol causes the symptoms associated with FASD.
Collapse
|
93
|
A novel cholinergic action of alcohol and the development of tolerance to that effect in Caenorhabditis elegans. Genetics 2014; 199:135-49. [PMID: 25342716 DOI: 10.1534/genetics.114.171884] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Understanding the genes and mechanisms involved in acute alcohol responses has the potential to allow us to predict an individual's predisposition to developing an alcohol use disorder. To better understand the molecular pathways involved in the activating effects of alcohol and the acute functional tolerance that can develop to such effects, we characterized a novel ethanol-induced hypercontraction response displayed by Caenorhabditis elegans. We compared body size of animals prior to and during ethanol treatment and showed that acute exposure to ethanol produced a concentration-dependent decrease in size followed by recovery to their untreated size by 40 min despite continuous treatment. An increase in cholinergic signaling, leading to muscle hypercontraction, is implicated in this effect because pretreatment with mecamylamine, a nicotinic acetylcholine receptor (nAChR) antagonist, blocked ethanol-induced hypercontraction, as did mutations causing defects in cholinergic signaling (cha-1 and unc-17). Analysis of mutations affecting specific subunits of nAChRs excluded a role for the ACR-2R, the ACR-16R, and the levamisole-sensitive AChR and indicated that this excitation effect is dependent on an uncharacterized nAChR that contains the UNC-63 α-subunit. We performed a forward genetic screen and identified eg200, a mutation that affects a conserved glycine in EAT-6, the α-subunit of the Na(+)/K(+) ATPase. The eat-6(eg200) mutant fails to develop tolerance to ethanol-induced hypercontraction and remains contracted for at least 3 hr of continuous ethanol exposure. These data suggest that cholinergic signaling through a specific α-subunit-containing nAChR is involved in ethanol-induced excitation and that tolerance to this ethanol effect is modulated by Na(+)/K(+) ATPase function.
Collapse
|
94
|
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describes a wide range of phenotypic defects affecting facial and neurological development associated with ethanol teratogenicity. It affects approximately 1 in 100 children born in the United States each year. Genetic predisposition along with timing and dosage of ethanol exposure are critical in understanding the prevalence and variability of FASD. The zebrafish attributes of external fertilization, genetic tractability, and high fecundity make it a powerful tool for FASD studies. However, a lack of consensus of ethanol treatment paradigms has limited the interpretation of these various studies. Here we address this concern by examining ethanol tissue concentrations across timing and genetic background. We utilize headspace gas chromatography to determine ethanol concentration in the AB, fli1:EGFP, and Tu backgrounds. In addition, we treated these embryos with ethanol over two different developmental time windows, 6-24 h post fertilization (hpf) and 24-48 hpf. Our analysis demonstrates that embryos rapidly equilibrate to a sub-media level of ethanol. Embryos then maintain this level of ethanol for the duration of exposure. The ethanol tissue concentration level is independent of genetic background, but is timing-dependent. Embryos exposed from 6 to 24 hpf were 2.7-4.2-fold lower than media levels, while embryos were 5.7-6.2-fold lower at 48 hpf. This suggests that embryos strengthen one or more barriers to ethanol as they develop. In addition, both the embryo and, to a lesser extent, the chorion, surrounding the embryo are barriers to ethanol. Overall, this work will help tighten ethanol treatment regimens and strengthen zebrafish as a model of FASD.
Collapse
Affiliation(s)
- C Ben Lovely
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA; Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX 78713, USA.
| | - Regina D Nobles
- Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Johann K Eberhart
- Molecular Biosciences, University of Texas at Austin, Austin, TX 78713, USA; Waggoner Center for Alcohol & Addiction Research, University of Texas at Austin, Austin, TX 78713, USA
| |
Collapse
|
95
|
Laurent AJ, Bindslev N, Johansson B, Berg L. Synergistic effects of ethanol and isopentenyl pyrophosphate on expansion of γδ T cells in synovial fluid from patients with arthritis. PLoS One 2014; 9:e103683. [PMID: 25090614 PMCID: PMC4121167 DOI: 10.1371/journal.pone.0103683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 04/29/2014] [Indexed: 01/03/2023] Open
Abstract
Low to moderate ethanol consumption has been associated with protective effects in autoimmune diseases such as rheumatoid arthritis, RA. An expansion of γδ T cells induced by isopentenyl pyrophosphate, IPP, likewise seems to have a protective role in arthritis. The aim of this project was to test the hypothesis that low doses of ethanol can enhance IPP-induced expansion of synovial fluid γδ T cells from patients with arthritis and may thereby potentially account for the beneficial effects of ethanol on symptoms of the arthritic process. Thus, mononuclear cells from synovial fluid (SF) from 15 patients with arthritis and from peripheral blood (PB) from 15 healthy donors were stimulated with low concentrations of ethanol and IPP for 7 days in vitro. IPP in combination with ethanol 0.015%, 2.5 mM, equivalent to the decrease per hour in blood ethanol concentration due to metabolism, gave a significantly higher fractional expansion of SF γδ T cells compared with IPP alone after 7 days (ratio 10.1+/-4.0, p<0.0008, n = 12) in patients with arthritis. Similar results were obtained for PB γδ T cells from healthy controls (ratio 2.0+/-0.4, p<0.011, n = 15). The augmented expansion of γδ T cells in SF is explained by a higher proliferation (p = 0.0034, n = 11) and an increased survival (p<0.005, n = 11) in SF cultures stimulated with IPP plus ethanol compared to IPP alone. The synergistic effects of IPP and ethanol indicate a possible allosteric effect of ethanol. Similar effects could be seen when stimulating PB with ethanol in presence of risedronate, which has the ability to increase endogenous levels of IPP. We conclude that expansion of γδ T cells by combinatorial drug effects, possibly in fixed-dose combination, FDC, of ethanol in the presence of IPP might give a protective role in diseases such as arthritis.
Collapse
MESH Headings
- Adult
- Aged
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Drug Synergism
- Ethanol/pharmacology
- Etidronic Acid/analogs & derivatives
- Etidronic Acid/pharmacology
- Etidronic Acid/therapeutic use
- Female
- Hemiterpenes/pharmacology
- Humans
- Interferon-gamma/biosynthesis
- Lymphocyte Activation/drug effects
- Male
- Middle Aged
- Models, Biological
- Organophosphorus Compounds/pharmacology
- Phenotype
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Risedronic Acid
- Synovial Fluid/cytology
- Synovial Fluid/drug effects
- Synovial Fluid/immunology
- T-Lymphocyte Subsets/drug effects
Collapse
Affiliation(s)
- Agneta J. Laurent
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Niels Bindslev
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Björn Johansson
- Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Louise Berg
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
96
|
Tarragon E, Baliño P, Aragon CMG. Centrally formed acetaldehyde mediates ethanol-induced brain PKA activation. Neurosci Lett 2014; 580:68-73. [PMID: 25093700 DOI: 10.1016/j.neulet.2014.07.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/28/2022]
Abstract
Centrally formed acetaldehyde has proven to be responsible for several psychopharmacological effects induced by ethanol. In addition, it has been suggested that the cAMP-PKA signaling transduction pathway plays an important role in the modulation of several ethanol-induced behaviors. Therefore, we hypothesized that acetaldehyde might be ultimately responsible for the activation of this intracellular pathway. We used three pharmacological agents that modify acetaldehyde activity (α-lipoic acid, aminotriazole, and d-penicillamine) to study the role of this metabolite on EtOH-induced PKA activation in mice. Our results show that the injection of α-lipoic acid, aminotriazole and d-penicillamine prior to acute EtOH administration effectively blocks the PKA-enhanced response to EtOH in the brain. These results strongly support the hypothesis of a selective release of acetaldehyde-dependent Ca(2+) as the mechanism involved in the neurobehavioral effects elicited by EtOH.
Collapse
Affiliation(s)
- E Tarragon
- Área de Psicobiología, Universitat Jaume I, Castellon de la Plana, Spain
| | - P Baliño
- Área de Psicobiología, Universitat Jaume I, Castellon de la Plana, Spain
| | - C M G Aragon
- Área de Psicobiología, Universitat Jaume I, Castellon de la Plana, Spain.
| |
Collapse
|
97
|
Trudell JR, Messing RO, Mayfield J, Harris RA. Alcohol dependence: molecular and behavioral evidence. Trends Pharmacol Sci 2014; 35:317-23. [PMID: 24865944 PMCID: PMC4089033 DOI: 10.1016/j.tips.2014.04.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/18/2014] [Accepted: 04/30/2014] [Indexed: 10/25/2022]
Abstract
Alcohol dependence is a complex condition with clear genetic factors. Some of the leading candidate genes code for subunits of the inhibitory GABAA and glycine receptors. These and related ion channels are also targets for the acute actions of alcohol, and there is considerable progress in understanding interactions of alcohol with these proteins at the molecular and even atomic levels. X-ray structures of open and closed states of ion channels combined with structural modeling and site-directed mutagenesis have elucidated direct actions of alcohol. Alcohol also alters channel function by translational and post-translational mechanisms, including phosphorylation and protein trafficking. Construction of mutant mice with either deletion of key proteins or introduction of alcohol-resistant channels has further linked specific proteins with discrete behavioral effects of alcohol. A combination of approaches, including genome wide association studies in humans, continues to advance the molecular basis of alcohol action on receptor structure and function.
Collapse
Affiliation(s)
- James R Trudell
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
98
|
An alcohol-sensing site in the calcium- and voltage-gated, large conductance potassium (BK) channel. Proc Natl Acad Sci U S A 2014; 111:9313-8. [PMID: 24927535 DOI: 10.1073/pnas.1317363111] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Ethanol alters BK (slo1) channel function leading to perturbation of physiology and behavior. Site(s) and mechanism(s) of ethanol-BK channel interaction are unknown. We demonstrate that ethanol docks onto a water-accessible site that is strategically positioned between the slo1 calcium-sensors and gate. Ethanol only accesses this site in presence of calcium, the BK channel's physiological agonist. Within the site, ethanol hydrogen-bonds with K361. Moreover, substitutions that hamper hydrogen bond formation or prevent ethanol from accessing K361 abolish alcohol action without altering basal channel function. Alcohol interacting site dimensions are approximately 10.7 × 8.6 × 7.1 Å, accommodating effective (ethanol-heptanol) but not ineffective (octanol, nonanol) channel activators. This study presents: (i) to our knowledge, the first identification and characterization of an n-alkanol recognition site in a member of the voltage-gated TM6 channel superfamily; (ii) structural insights on ethanol allosteric interactions with ligand-gated ion channels; and (iii) a first step for designing agents that antagonize BK channel-mediated alcohol actions without perturbing basal channel function.
Collapse
|
99
|
Maguire EP, Mitchell EA, Greig SJ, Corteen N, Balfour DJK, Swinny JD, Lambert JJ, Belelli D. Extrasynaptic glycine receptors of rodent dorsal raphe serotonergic neurons: a sensitive target for ethanol. Neuropsychopharmacology 2014; 39:1232-44. [PMID: 24264816 PMCID: PMC3957119 DOI: 10.1038/npp.2013.326] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 10/17/2013] [Accepted: 11/12/2013] [Indexed: 11/08/2022]
Abstract
Alcohol abuse is a significant medical and social problem. Several neurotransmitter systems are implicated in ethanol's actions, with certain receptors and ion channels emerging as putative targets. The dorsal raphe (DR) nucleus is associated with the behavioral actions of alcohol, but ethanol actions on these neurons are not well understood. Here, using immunohistochemistry and electrophysiology we characterize DR inhibitory transmission and its sensitivity to ethanol. DR neurons exhibit inhibitory 'phasic' post-synaptic currents mediated primarily by synaptic GABAA receptors (GABAAR) and, to a lesser extent, by synaptic glycine receptors (GlyR). In addition to such phasic transmission mediated by the vesicular release of neurotransmitter, the activity of certain neurons may be governed by a 'tonic' conductance resulting from ambient GABA activating extrasynaptic GABAARs. However, for DR neurons extrasynaptic GABAARs exert only a limited influence. By contrast, we report that unusually the GlyR antagonist strychnine reveals a large tonic conductance mediated by extrasynaptic GlyRs, which dominates DR inhibition. In agreement, for DR neurons strychnine increases their input resistance, induces membrane depolarization, and consequently augments their excitability. Importantly, this glycinergic conductance is greatly enhanced in a strychnine-sensitive fashion, by behaviorally relevant ethanol concentrations, by drugs used for the treatment of alcohol withdrawal, and by taurine, an ingredient of certain 'energy drinks' often imbibed with ethanol. These findings identify extrasynaptic GlyRs as critical regulators of DR excitability and a novel molecular target for ethanol.
Collapse
Affiliation(s)
- Edward P Maguire
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Elizabeth A Mitchell
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Scott J Greig
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Nicole Corteen
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - David J K Balfour
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, Dundee University, Dundee, UK
| |
Collapse
|
100
|
Silveri MM. GABAergic contributions to alcohol responsivity during adolescence: insights from preclinical and clinical studies. Pharmacol Ther 2014; 143:197-216. [PMID: 24631274 DOI: 10.1016/j.pharmthera.2014.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 02/28/2014] [Indexed: 01/04/2023]
Abstract
There is a considerable body of literature demonstrating that adolescence is a unique age period, which includes rapid and dramatic maturation of behavioral, cognitive, hormonal and neurobiological systems. Most notably, adolescence is also a period of unique responsiveness to alcohol effects, with both hyposensitivity and hypersensitivity observed to the various effects of alcohol. Multiple neurotransmitter systems are undergoing fine-tuning during this critical period of brain development, including those that contribute to the rewarding effects of drugs of abuse. The role of developmental maturation of the γ-amino-butyric acid (GABA) system, however, has received less attention in contributing to age-specific alcohol sensitivities. This review integrates GABA findings from human magnetic resonance spectroscopy studies as they may translate to understanding adolescent-specific responsiveness to alcohol effects. Better understanding of the vulnerability of the GABA system both during adolescent development, and in psychiatric conditions that include alcohol dependence, could point to a putative mechanism, boosting brain GABA, that may have increased effectiveness for treating alcohol use disorders.
Collapse
Affiliation(s)
- Marisa M Silveri
- Neurodevelopmental Laboratory on Addictions and Mental Health, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|