51
|
Pervin Z, Stephen JM. Effect of alcohol on the central nervous system to develop neurological disorder: pathophysiological and lifestyle modulation can be potential therapeutic options for alcohol-induced neurotoxication. AIMS Neurosci 2021; 8:390-413. [PMID: 34183988 PMCID: PMC8222771 DOI: 10.3934/neuroscience.2021021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/01/2021] [Indexed: 12/06/2022] Open
Abstract
The central nervous system (CNS) is the major target for adverse effects of alcohol and extensively promotes the development of a significant number of neurological diseases such as stroke, brain tumor, multiple sclerosis (MS), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS). Excessive alcohol consumption causes severe neuro-immunological changes in the internal organs including irreversible brain injury and it also reacts with the defense mechanism of the blood-brain barrier (BBB) which in turn leads to changes in the configuration of the tight junction of endothelial cells and white matter thickness of the brain. Neuronal injury associated with malnutrition and oxidative stress-related BBB dysfunction may cause neuronal degeneration and demyelination in patients with alcohol use disorder (AUD); however, the underlying mechanism still remains unknown. To address this question, studies need to be performed on the contributing mechanisms of alcohol on pathological relationships of neurodegeneration that cause permanent neuronal damage. Moreover, alcohol-induced molecular changes of white matter with conduction disturbance in neurotransmission are a likely cause of myelin defect or axonal loss which correlates with cognitive dysfunctions in AUD. To extend our current knowledge in developing a neuroprotective environment, we need to explore the pathophysiology of ethanol (EtOH) metabolism and its effect on the CNS. Recent epidemiological studies and experimental animal research have revealed the association between excessive alcohol consumption and neurodegeneration. This review supports an interdisciplinary treatment protocol to protect the nervous system and to improve the cognitive outcomes of patients who suffer from alcohol-related neurodegeneration as well as clarify the pathological involvement of alcohol in causing other major neurological disorders.
Collapse
Affiliation(s)
- Zinia Pervin
- Department of Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | - Julia M Stephen
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM 87106, USA
| |
Collapse
|
52
|
Martínez-Magaña JJ, Genis-Mendoza AD, Villatoro Velázquez JA, Bustos-Gamiño M, Juárez-Rojop IE, Tovilla-Zarate CA, Sarmiento E, Saucedo E, Rodríguez-Mayoral O, Fleiz-Bautista C, Camarena B, Aguilar A, Gonzalez-Castro TB, Medina-Mora ME, Nicolini H. Genome-wide association study of psychiatric and substance use comorbidity in Mexican individuals. Sci Rep 2021; 11:6771. [PMID: 33762635 PMCID: PMC7990941 DOI: 10.1038/s41598-021-85881-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/08/2021] [Indexed: 01/31/2023] Open
Abstract
The combination of substance use and psychiatric disorders is one of the most common comorbidities. The objective of this study was to perform a genome-wide association study of this comorbidity (Com), substance use alone (Subs), and psychiatric symptomatology alone (Psych) in the Mexican population. The study included 3914 individuals of Mexican descent. Genotyping was carried out using the PsychArray microarray and genome-wide correlations were calculated. Genome-wide associations were analyzed using multiple logistic models, polygenic risk scores (PRSs) were evaluated using multinomial models, and vertical pleiotropy was evaluated by generalized summary-data-based Mendelian randomization. Brain DNA methylation quantitative loci (brain meQTL) were also evaluated in the prefrontal cortex. Genome-wide correlation and vertical pleiotropy were found between all traits. No genome-wide association signals were found, but 64 single-nucleotide polymorphism (SNPs) reached nominal associations (p < 5.00e-05). The SNPs associated with each trait were independent, and the individuals with high PRSs had a higher prevalence of tobacco and alcohol use. In the multinomial models all of the PRSs (Subs-PRS, Com-PRS, and Psych-PRS) were associated with all of the traits. Brain meQTL of the Subs-associated SNPs had an effect on the genes enriched in insulin signaling pathway, and that of the Psych-associated SNPs had an effect on the Fc gamma receptor phagocytosis pathway.
Collapse
Affiliation(s)
- José Jaime Martínez-Magaña
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, Mexico
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Alma Delia Genis-Mendoza
- Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Hospital Psiquiátrico Infantil Juan N. Navarro, Servicios de Atención Psiquiátrica, Mexico City, Mexico
| | - Jorge Ameth Villatoro Velázquez
- Unidad de Encuestas y Análisis de Datos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
- Seminario de Estudios Sobre la Globalidad, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City, Mexico
| | - Marycarmen Bustos-Gamiño
- Unidad de Encuestas y Análisis de Datos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
| | - Isela Esther Juárez-Rojop
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, Mexico
| | | | - Emmanuel Sarmiento
- Hospital Psiquiátrico Infantil Juan N. Navarro, Servicios de Atención Psiquiátrica, Mexico City, Mexico
| | - Erasmo Saucedo
- Centro de Neurociencias Avanzadas, Departamento de Psiquiátrica del Hospital Psiquiátrico, Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | | | - Clara Fleiz-Bautista
- Unidad de Encuestas y Análisis de Datos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
- Seminario de Estudios Sobre la Globalidad, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City, Mexico
| | - Beatriz Camarena
- Laboratorio de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
| | - Alejandro Aguilar
- Laboratorio de Farmacogenética, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
| | - Thelma Beatriz Gonzalez-Castro
- División Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Jalpa de Méndez, Tabasco, Mexico
| | - María Elena Medina-Mora
- Unidad de Encuestas y Análisis de Datos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz (INPRFM), Mexico City, Mexico
- Seminario de Estudios Sobre la Globalidad, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City, Mexico
| | - Humberto Nicolini
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, Mexico.
- Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan, 14610, Mexico City, Mexico.
| |
Collapse
|
53
|
Castro‐Zavala A, Martín‐Sánchez A, Luján MÁ, Valverde O. Maternal separation increases cocaine intake through a mechanism involving plasticity in glutamate signalling. Addict Biol 2021; 26:e12911. [PMID: 32329565 DOI: 10.1111/adb.12911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Early-life stress (ELS) is associated with negative consequences, including maladaptive long-lasting brain effects. These alterations seem to increase the likelihood of developing substance use disorders. However, the molecular consequences of ELS are poorly understood. In the present study, we tested the impact of ELS induced by maternal separation with early weaning (MSEW) in CD1 male mice at different phases of cocaine self-administration (SA). We also investigated the subsequent alterations on GluR2, GluR1, cAMP response element-binding (CREB), and CREB-phosphorylation (pCREB) in ventral tegmental area (VTA) and nucleus accumbens (NAc) induced by both MSEW and cocaine SA. Our results show that MSEW animals expressed a higher cocaine intake, an increased vulnerability to the acquisition of cocaine SA, and incapacity to extinguish cocaine SA behaviour. MSEW mice showed decreased GluR2 and increased GluR1 and pCREB in NAc. Also, results displayed reduction of basal levels of GluR1 and CREB and an elevation of GluR1/GluR2 ratio in the VTA. Such results hint at an enhanced glutamatergic function in NAc and increased excitability of VTA DA neurons in maternally separated mice. Altogether, our results suggest that MSEW induces molecular alterations in the brain areas related to reward processing, increasing the vulnerability to depression and cocaine-seeking behaviour.
Collapse
Affiliation(s)
- Adriana Castro‐Zavala
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
| | - Ana Martín‐Sánchez
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
- Neuroscience Research Programme IMIM‐Hospital del Mar Research Institute Barcelona Spain
| | - Miguel Ángel Luján
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
- Neuroscience Research Programme IMIM‐Hospital del Mar Research Institute Barcelona Spain
| |
Collapse
|
54
|
Kahvandi N, Ebrahimi Z, Karimi SA, Shahidi S, Salehi I, Naderishahab M, Sarihi A. The effect of the mGlu8 receptor agonist, (S)-3,4-DCPG on acquisition and expression of morphine-induced conditioned place preference in male rats. Behav Brain Funct 2021; 17:1. [PMID: 33612106 PMCID: PMC7897377 DOI: 10.1186/s12993-021-00174-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/05/2021] [Indexed: 02/24/2023] Open
Abstract
Background The nucleus accumbens (NAc) plays a principal role in drug reward. It has been reported that metabotropic glutamate receptors (mGlu receptors) play a key role in the rewarding pathway(s). Previous studies have shown the vast allocation of the different types of mGlu receptors, including mGlu8 receptors, in regions that are associated with opioid rewards, such as the NAc. The aim of the present study was to evaluate the role of mGlu8 receptors within the NAc in the acquisition and expression phases of morphine induced conditioned place preference (CPP). Adult male Wistar rats were bilaterally implanted by two cannulas' in the NAc and were evaluated in a CPP paradigm. Selective mGlu8 receptor allosteric agonist (S-3,4-DCPG) was administered at doses of 0.03, 0.3, and 3 μg/0.5 μL saline per side into the NAc on both sides during the 3 days of morphine (5 mg/kg) conditioning (acquisition) phase, or before place preference test, or post-conditioning (expression) phase of morphine-induced CPP. Results The results revealed that intra-accumbal administration of S-3,4-DCPG (0.3 and 3 μg) markedly decreased the acquisition in a dose-dependent manner but had no effect on expression of morphine-induced CPP. Conclusions The findings suggest that activation of mGlu8 receptors in the NAc dose-dependently blocks the establishment of morphine-induced CPP and reduces the rewarding properties of morphine which may be related to the glutamate activity into the NAc and in reward pathway(s). These data suggest that mGlu8 receptor may be involved in conditioned morphine reward.
Collapse
Affiliation(s)
- Nazanin Kahvandi
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Zahra Ebrahimi
- Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran.,Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran.,Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran.,Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Naderishahab
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, Iran. .,Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
55
|
Oliveira TPD, Gonçalves BDC, Oliveira BS, de Oliveira ACP, Reis HJ, Ferreira CN, Aguiar DC, de Miranda AS, Ribeiro FM, Vieira EML, Palotás A, Vieira LB. Negative Modulation of the Metabotropic Glutamate Receptor Type 5 as a Potential Therapeutic Strategy in Obesity and Binge-Like Eating Behavior. Front Neurosci 2021; 15:631311. [PMID: 33642987 PMCID: PMC7902877 DOI: 10.3389/fnins.2021.631311] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is a multifactorial disease, which in turn contributes to the onset of comorbidities, such as diabetes and atherosclerosis. Moreover, there are only few options available for treating obesity, and most current pharmacotherapy causes severe adverse effects, while offering minimal weight loss. Literature shows that metabotropic glutamate receptor 5 (mGluR5) modulates central reward pathways. Herein, we evaluated the effect of VU0409106, a negative allosteric modulator (NAM) of mGluR5 in regulating feeding and obesity parameters. Diet-induced obese C57BL/6 mice were treated for 14 days with VU0409106, and food intake, body weight, inflammatory/hormonal levels, and behavioral tests were performed. Our data suggest reduction of feeding, body weight, and adipose tissue inflammation in mice treated with high-fat diet (HFD) after chronic treatment with VU0409106. Furthermore, a negative modulation of mGluR5 also reduces binge-like eating, the most common type of eating disorder. Altogether, our results pointed out mGluR5 as a potential target for treating obesity, as well as related disorders.
Collapse
Affiliation(s)
- Tadeu P. D. Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno D. C. Gonçalves
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruna S. Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antonio Carlos P. de Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton J. Reis
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Claudia N. Ferreira
- Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniele C. Aguiar
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline S. de Miranda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiola M. Ribeiro
- Departamento de Bioquimica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erica M. L. Vieira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - András Palotás
- Asklepios-Med (Private Medical Practice and Research Center), Szeged, Hungary
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Luciene B. Vieira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
56
|
Szpręgiel I, Wrońska D, Kmiecik M, Pałka S, Kania BF. Glutamic Acid Decarboxylase Concentration Changes in Response to Stress and Altered Availability of Glutamic Acid in Rabbit ( Oryctolagus cuniculus) Brain Limbic Structures. Animals (Basel) 2021; 11:455. [PMID: 33572286 PMCID: PMC7915518 DOI: 10.3390/ani11020455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
Glutamic acid decarboxylase (GAD) is an enzyme that catalyses the formation of γ-aminobutyric acid (GABA), the most important inhibitory neurotransmitter, from glutamic acid (Glu), which is considered the most important excitatory transmitter in the central and peripheral nervous systems. GAD is a key enzyme that provides a balance between Glu and GABA concentration. Hence, it can be assumed that if the GAD executes the synthesis of GABA from Glu, it is important in the stress response, and thus also in triggering the emotional states of the body that accompany stress. The aim of the study was to investigate the concentration of the GAD in motivational structures in the brain of the rabbit (Oryctolagus cuniculus) under altered homeostatic conditions caused by stress and variable availability of Glu. Summarising, the experimental results clearly showed variable concentrations of GAD in the motivational structures of the rabbit brain. The highest concentration of GAD was found in the hypothalamus, which suggests a strong effect of Glu and GABA on the activity of this brain structure. The GAD concentrations in individual experimental groups depended to a greater extent on blocking the activity of glutamate receptors than on the effects of a single stress exposure. The results obtained clearly support the possibility that a rapid change in the concentration of GAD could shift bodily responses to quickly achieve homeostasis, especially in this species. Further studies are necessary to reveal the role of the Glu-GAD-GABA system in the modulation of stress situations as well as in body homeostasis.
Collapse
Affiliation(s)
- Izabela Szpręgiel
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Danuta Wrońska
- Department of Animal Physiology and Endocrinology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland;
| | - Michał Kmiecik
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Sylwia Pałka
- Department of Genetics, Animal Breeding and Ethology, Faculty of Animal Sciences, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Kraków, Poland; (M.K.); (S.P.)
| | - Bogdan F. Kania
- University Centre of Veterinary Medicine JU-AU, University of Agriculture in Kraków, Mickiewicza 24/28, 30-059 Kraków, Poland;
| |
Collapse
|
57
|
Tamura EK, Oliveira-Silva KS, Ferreira-Moraes FA, Marinho EAV, Guerrero-Vargas NN. Circadian rhythms and substance use disorders: A bidirectional relationship. Pharmacol Biochem Behav 2021; 201:173105. [PMID: 33444601 DOI: 10.1016/j.pbb.2021.173105] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 01/23/2023]
Abstract
The circadian system organizes circadian rhythms (biological cycles that occur around 24 h) that couple environmental cues (zeitgebers) with internal functions of the organism. The misalignment between circadian rhythms and external cues is known as chronodisruption and contributes to the development of mental, metabolic and other disorders, including cancer, cardiovascular diseases and addictive disorders. Drug addiction represents a global public health concern and affects the health and well-being of individuals, families and communities. In this manuscript, we reviewed evidence indicating a bidirectional relationship between the circadian system and the development of addictive disorders. We provide information on the interaction between the circadian system and drug addiction for each drug or drug class (alcohol, cannabis, hallucinogens, psychostimulants and opioids). We also describe evidence showing that drug use follows a circadian pattern, which changes with the progression of addiction. Furthermore, clock gene expression is also altered during the development of drug addiction in many brain areas related to drug reward, drug seeking and relapse. The regulation of the glutamatergic and dopaminergic neurocircuitry by clock genes is postulated to be the main circadian mechanism underlying the escalation of drug addiction. The bidirectional interaction between the circadian system and drug addiction seems to be mediated by the effects caused by each drug or class of drugs of abuse. These studies provide new insights on the development of successful strategies aimed at restoring/stabilizing circadian rhythms to reduce the risk for addiction development and relapse.
Collapse
Affiliation(s)
- Eduardo K Tamura
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil.
| | - Kallyane S Oliveira-Silva
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Felipe A Ferreira-Moraes
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Eduardo A V Marinho
- Department of Health Sciences, Universidade Estadual de Santa Cruz, BR-415, Rodovia Ilhéus- Itabuna, Km-16, Salobrinho, Ilhéus, Bahia 45662-000, Brazil
| | - Natalí N Guerrero-Vargas
- Department of Anatomy, Faculty of Medicine, Universidad Nacional Autonóma de México, Av Universidad 3000, Ciudad Universitaria, México City 04510, Mexico
| |
Collapse
|
58
|
Wang G, Weber-Fahr W, Frischknecht U, Hermann D, Kiefer F, Ende G, Sack M. Cortical Glutamate and GABA Changes During Early Abstinence in Alcohol Dependence and Their Associations With Benzodiazepine Medication. Front Psychiatry 2021; 12:656468. [PMID: 34290627 PMCID: PMC8287125 DOI: 10.3389/fpsyt.2021.656468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
In this report, we present cross-sectional and longitudinal findings from single-voxel MEGA-PRESS MRS of GABA as well as Glu, and Glu + glutamine (Glx) concentrations in the ACC of treatment-seeking alcohol-dependent patients (ADPs) during detoxification (first 2 weeks of abstinence). The focus of this study was to examine whether the amount of benzodiazepine administered to treat withdrawal symptoms was associated with longitudinal changes in Glu, Glx, and GABA. The tNAA levels served as an internal quality reference; in agreement with the vast majority of previous reports, these levels were initially decreased and normalized during the course of abstinence in ADPs. Our results on Glu and Glx support hyperglutamatergic functioning during alcohol withdrawal, by showing higher ACC Glu and Glx levels on the first day of detoxification in ADPs. Withdrawal severity is reflected in cumulative benzodiazepine requirements throughout the withdrawal period. The importance of withdrawal severity for the study of GABA and Glu changes in early abstinence is emphasized by the benzodiazepine-dependent Glu, Glx, and GABA changes observed during the course of abstinence.
Collapse
Affiliation(s)
- Guoying Wang
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Ulrich Frischknecht
- Department of Addiction Medicine and Addictive Behavior, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany.,German Institute of Addiction and Prevention Research, Catholic University of Applied Sciences, Cologne, Germany
| | - Derik Hermann
- Department of Addiction Medicine and Addictive Behavior, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany.,Therapieverbund Ludwigsmühle, Landau in der Pfalz, Germany
| | - Falk Kiefer
- Department of Addiction Medicine and Addictive Behavior, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Gabriele Ende
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Markus Sack
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
59
|
De Sa Nogueira D, Bourdy R, Filliol D, Quessada C, McCort-Tranchepain I, Acher F, Zwiller J, Romieu P, Befort K. LSP2-9166, an orthosteric mGlu4 and mGlu7 receptor agonist, reduces cocaine self-administration under a progressive ratio schedule in rats. Neurosci Lett 2020; 764:135603. [PMID: 33387661 DOI: 10.1016/j.neulet.2020.135603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 11/24/2022]
Abstract
Cocaine addiction is a serious health issue in Western countries. Despite the regular increase in cocaine consumption across the population, there is no specific treatment for cocaine addiction. Critical roles for glutamate neurotransmission in the rewarding effects of psychostimulants as well as relapse have been suggested and accumulating evidence indicates that targeting mGlu group III receptors could represent a promising strategy to develop therapeutic compounds to treat addiction. In this context, the aim of our study was to examine the effect of LSP2-9166, a mGlu4/mGlu7 receptor orthosteric agonist, on the motivation for cocaine intake. We used an intravenous self-administration paradigm in male Wistar rats as a reliable model of voluntary drug intake. We first evaluated the direct impact of cocaine on Grm4 and Grm7 gene expression. Voluntary cocaine intake under a fixed ratio schedule of injections induced an increase of both mGlu4 and mGlu7 receptor transcripts in nucleus accumbens and hippocampus. We then evaluated the ability of LSP2-9166 to affect cocaine self-administration under a progressive ratio schedule of reinforcement. We found that this compound inhibits the motivation to obtain the drug, although it induced a hypolocomotor effect which could biais motivation index. Our findings demonstrate that mGlu group III receptors represent new targets for decreasing motivation to self-administer cocaine.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Romain Bourdy
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Dominique Filliol
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France; Université de Strasbourg, Inserm, UMR-S1118, 11 rue Humann, F-67000, Strasbourg, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université de Paris 45 rue des Saints-Pères, F-75270, Paris Cedex 6, France
| | - Francine Acher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université de Paris 45 rue des Saints-Pères, F-75270, Paris Cedex 6, France
| | - Jean Zwiller
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Pascal Romieu
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France.
| |
Collapse
|
60
|
Stratilov VA, Tyulkova EI, Vetrovoy OV. Prenatal Stress as a Factor of the
Development of Addictive States. J EVOL BIOCHEM PHYS+ 2020. [DOI: 10.1134/s0022093020060010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
61
|
Enduring dysregulation of nucleus accumbens catecholamine and glutamate transmission by developmental exposure to phenylpropanolamine. Brain Res 2020; 1748:147098. [PMID: 32896521 DOI: 10.1016/j.brainres.2020.147098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 11/24/2022]
Abstract
For over 50 years, the sympathomimetic phenylpropanolamine (PPA; ±-norephedrine) was a primary active ingredient in over-the-counter nasal decongestants for both children and adults and continues to be prevalent in the vast majority of countries today. Previously, we reported that juvenile PPA exposure alters the developmental trajectory of catecholamine and amino acid neurotransmitter systems in the nucleus accumbens (NAC), impacting the motivational valence of cocaine in later life. The present study employed a combination of in vivo microdialysis and immunoblotting approaches to better understand how juvenile PPA exposure impacts catecholamine and glutamate function within the NAC. For this, C57BL/6J mice were pretreated repeatedly with PPA (0 or 40 mg/kg) during postnatal days 21-33. Starting at 70 days of age, the function and expression of receptors and transporters regulating extracellular dopamine and glutamate were determined. Juvenile PPA pretreatment completely abolished the capacity of selective dopamine and epinephrine reuptake inhibitors to increase NAC levels of both catecholamines, without impacting D2 or α2 receptor regulation of catecholamine release. Juvenile PPA pretreatment facilitated the rise in NAC glutamate elicited by dopamine, norepinephrine and glutamate transporter inhibitors and blunted mGlu2/3 inhibition of glutamate release in this region. These data confirm that juvenile exposure to PPA produces protracted perturbations in the regulation of extracellular catecholamine and glutamate levels within the NAC and further the hypothesis that early exposure to sympathomimetic drugs found in cough, cold and allergy medicines, have long-lasting effects upon neurotransmission within brain regions gating motivation.
Collapse
|
62
|
Vaseghi S, Nasehi M, Zarrindast MR. How do stupendous cannabinoids modulate memory processing via affecting neurotransmitter systems? Neurosci Biobehav Rev 2020; 120:173-221. [PMID: 33171142 DOI: 10.1016/j.neubiorev.2020.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 12/27/2022]
Abstract
In the present study, we wanted to review the role of cannabinoids in learning and memory in animal models, with respect to their interaction effects with six principal neurotransmitters involved in learning and memory including dopamine, glutamate, GABA (γ-aminobutyric acid), serotonin, acetylcholine, and noradrenaline. Cannabinoids induce a wide-range of unpredictable effects on cognitive functions, while their mechanisms are not fully understood. Cannabinoids in different brain regions and in interaction with different neurotransmitters, show diverse responses. Previous findings have shown that cannabinoids agonists and antagonists induce various unpredictable effects such as similar effect, paradoxical effect, or dualistic effect. It should not be forgotten that brain neurotransmitter systems can also play unpredictable roles in mediating cognitive functions. Thus, we aimed to review and discuss the effect of cannabinoids in interaction with neurotransmitters on learning and memory. In addition, we mentioned to the type of interactions between cannabinoids and neurotransmitter systems. We suggested that investigating the type of interactions is a critical neuropharmacological issue that should be considered in future studies.
Collapse
Affiliation(s)
- Salar Vaseghi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
63
|
The infralimbic cortex and mGlu5 mediate the effects of chronic intermittent ethanol exposure on fear learning and memory. Psychopharmacology (Berl) 2020; 237:3417-3433. [PMID: 32767063 PMCID: PMC7572878 DOI: 10.1007/s00213-020-05622-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/27/2020] [Indexed: 01/19/2023]
Abstract
RATIONALE AND OBJECTIVES Alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) often occur comorbidly. While the incidence of these disorders is increasing, there is little investigation into the interacting neural mechanisms between these disorders. These studies aim to identify cognitive deficits that occur as a consequence of fear and ethanol exposure, implement a novel pharmaceutical intervention, and determine relevant underlying neurocircuitry. Additionally, due to clinical sex differences in PTSD prevalence and alcohol abuse, these studies examine the nature of this relationship in rodent models. METHODS Animals were exposed to a model of PTSD+AUD using auditory fear conditioning followed by chronic intermittent ethanol exposure (CIE). Then, rats received extinction training consisting of multiple conditioned stimulus presentations in absence of the shock. Extinction recall and context-induced freezing were measured in subsequent tests. CDPPB, a metabotropic glutamate receptor 5 (mGlu5) positive allosteric modulator, was used to treat these deficits, and region-specific effects were determined using microinjections. RESULTS These studies determined that CIE exposure led to deficits in fear extinction learning and heightened context-induced freezing while sex differences emerged in fear conditioning and extinction cue recall tests. Furthermore, using CDPPB, these studies found that enhancement of infralimbic (IfL) mGlu5 activity was able to recover CIE-induced deficits in both males and females. CONCLUSIONS These studies show that CIE induces deficits in fear-related behaviors and that enhancement of IfL glutamatergic activity can facilitate learning during extinction. Additionally, we identify novel pharmacological targets for the treatment of individuals who suffer from PTSD and AUD.
Collapse
|
64
|
Rangel-Barajas C, Coronel I, Zhang Y, Hernández M, Boehm Ii SL. Low-level developmental lead exposure does not predispose to adult alcohol self-administration, but does increase the risk of relapsing to alcohol seeking in mice: Contrasting role of GLT1 and xCT brain expression. Neuropharmacology 2020; 181:108339. [PMID: 33010299 DOI: 10.1016/j.neuropharm.2020.108339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/27/2022]
Abstract
Lead (Pb) is a neurotoxic heavy metal pollutant. Despite the efforts to reduce Pb environmental exposure and to prevent Pb poisoning, exposure in human populations persists. Studies of adults with history of childhood lead exposure have consistently demonstrated cognitive impairments that have been associated with sustained glutamate signaling. Additionally, some clinical studies have also found correlations between Pb exposure and increased proclivity to drug addiction. Thus, here we sought to investigate if developmental Pb exposure can increase propensity to alcohol consumption and relapse using an alcohol self-administration paradigm. Because Pb exposure is associated with increased glutamatergic tone, we also studied the effects on the expression of synaptic and non-synaptic glutamate transporters in brain regions associated with drug addiction such as the nucleus accumbens (NAc), dorsomedial striatum (DMS), dorsolateral striatum (DLS), and medial prefrontal cortex (mPFC). We found that while developmental Pb exposure did not increase risk for alcohol self-administration, it did play a role in relapsing to alcohol. The effects were associated with differential expression of the glutamate transporter 1 (GLT1) and the glutamate/cystine antiporter (xCT). In the NAc and DLS the expression of GLT1 was found to be significantly reduced, while no changes were found in DMS or mPFC. Contrastingly, xCT was found to be upregulated in NAc but downregulated in DLS, with no changes in DMS or mPFC. Our data suggest that lead exposure is involved in relapse to alcohol seeking, an effect that could be associated with downregulation of GLT1 and xCT in the DLS.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Israel Coronel
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Yanping Zhang
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Maribel Hernández
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA
| | - Stephen L Boehm Ii
- Department of Psychology, School of Science, Indiana University-Purdue University Indianapolis, 402 N Blackford St, Indianapolis, IN, 46202, USA; Indiana Alcohol Research Center, Indiana University School of Medicine, 340 W 10th St, Indianapolis, IN, 462020, USA.
| |
Collapse
|
65
|
Gibson AS, Keefe KA, Furlong TM. Accelerated habitual learning resulting from L-dopa exposure in rats is prevented by N-acetylcysteine. Pharmacol Biochem Behav 2020; 198:173033. [PMID: 32888972 DOI: 10.1016/j.pbb.2020.173033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
Instrumental actions are initially goal-directed and driven by their associated outcome. However, with repeated experience habitual actions develop which are automated and efficient, as they are instead driven by antecedent stimuli. Dopamine is thought to facilitate the transition from goal-directed to habitual actions. This idea has been largely derived from evidence that psychostimulants accelerate the development of habitual actions. In the current study, we examined the impact of L-dopa (levodopa or L-dihydroxyphenylalanine), which also potentiates dopamine activity, on habitual learning. L-dopa was systemically administered prior to training rats to press a lever for a food outcome. When tested, L-dopa exposed animals were insensitive to changes in the value of the food outcome, and hence demonstrated accelerated habitual behavioral control compared to control animals that remained goal directed. We also showed that when N-acetylcysteine (NAC), an antioxidant and regulator of glutamate activity, was co-administered with L-dopa, it prevented the transition to habitual behavior; an effect demonstrated previously for cocaine. Therefore, this study establishes similarities between L-dopa and psychostimulants in both the development and prevention of habitual actions, and supports the notion that excess dopamine potentiates habitual learning. This finding extends the limited existing knowledge of the impact of L-dopa on learning and behavior, and has implications for neurological disorders where L-dopa is the primary treatment.
Collapse
Affiliation(s)
- Anne S Gibson
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA
| | - Kristen A Keefe
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA
| | - Teri M Furlong
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA; Neuroscience Research Australia, 139 Barker Street, Randwick, NSW, Australia; School of Medical Sciences, University of New South Wales, Kensington, NSW, Australia.
| |
Collapse
|
66
|
Li J, Wang H, Li M, Shen Q, Li X, Rong X, Peng Y. Efficacy of pharmacotherapeutics for patients comorbid with alcohol use disorders and depressive symptoms-A bayesian network meta-analysis. CNS Neurosci Ther 2020; 26:1185-1197. [PMID: 32686291 PMCID: PMC7564195 DOI: 10.1111/cns.13437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/25/2020] [Accepted: 06/28/2020] [Indexed: 12/05/2022] Open
Abstract
Background We aimed to compare and rank the efficacy of different pharmacotherapeutics for patients comorbid with alcohol use disorders and depressive symptoms. Method Bayesian network meta‐analysis was performed for three different outcome parameters: alcohol use disorders (AUD) remission rate, percent abstinent days, and scores of depression scales. The surface under the cumulative ranking curves (SUCRA) was used for ranking the efficacy of interventions. Sensitivity analysis and direct pairwise analysis were conducted to validate the main results. Results A total of 68 RCTs consisting of 5890 patients were included. Disulfiram could significantly increase the AUD remission rates (OR 5.02, 1.97‐12.95) and the percent abstinent days (MD 17.08, 3.48‐30.93). Disulfiram was associated with the best efficacy in achieving remission (SUCRA 95.1%) and increasing abstinent days (SUCRA 87.6%). Noradrenaline reuptake inhibitor was significantly more efficacious than controls (SMD −2.44, −3.53 to −1.36) and have the first rank (SUCRA 99.0%) in reducing the scores of depression scales. Antiepileptics have relatively higher ranks in efficacy for both AUD and depressive symptoms. Conclusions Disulfiram was associated with the best efficacy in achieving abstinence for comorbidity patients. Noradrenaline reuptake inhibitor was demonstrated to be associated with the best efficacy in reducing scores of depression scales. Antiepileptics might be beneficial to both alcohol‐related and depressive symptoms.
Collapse
Affiliation(s)
- Jiande Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongxuan Wang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mei Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiangpen Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ying Peng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
67
|
You JC, Muralidharan K, Fu CH, Park J, Tosi U, Zhang X, Chin J. Distinct patterns of dentate gyrus cell activation distinguish physiologic from aberrant stimuli. PLoS One 2020; 15:e0232241. [PMID: 32407421 PMCID: PMC7224541 DOI: 10.1371/journal.pone.0232241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 04/10/2020] [Indexed: 11/20/2022] Open
Abstract
Under physiologic conditions, the dentate gyrus (DG) exhibits exceptionally low levels of activity compared to other brain regions. A sparse activation pattern is observed even when the DG is engaged to process new information; for example, only ~1–3% of neurons in the DG granule cell layer (GCL) are activated after placing animals in a novel, enriched environment. Moreover, such physiologic stimulation of GCL neurons recruits young granule cells more readily than older cells. This sparse pattern of cell activation has largely been attributed to intrinsic circuit properties of the DG, such as reduced threshold for activation in younger cells, and increased inhibition onto older cells. Given these intrinsic properties, we asked whether such activation of young granule cells was unique to physiologic stimulation, or could be elicited by general pharmacological activation of the hippocampus. We found that administration of kainic acid (KA) at a low dose (5 mg/kg) to wildtype C57BL/6 mice activated a similarly sparse number of cells in the GCL as physiologic DG stimulation by exposure to a novel, enriched environment. However, unlike physiologic stimulation, 5 mg/kg KA activated primarily old granule cells as well as GABAergic interneurons. This finding indicates that intrinsic circuit properties of the DG alone may not be sufficient to support the engagement of young granule cells, and suggest that other factors such as the specificity of the pattern of inputs, may be involved.
Collapse
Affiliation(s)
- Jason C. You
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kavitha Muralidharan
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Chia-Hsuan Fu
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jin Park
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Umberto Tosi
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Xiaohong Zhang
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jeannie Chin
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Memory & Brain Research Center, Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
68
|
Behavioral plasticity and gene regulation in the brain during an intermittent ethanol exposure in adult zebrafish population. Pharmacol Biochem Behav 2020; 192:172909. [DOI: 10.1016/j.pbb.2020.172909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/27/2020] [Accepted: 03/12/2020] [Indexed: 01/04/2023]
|
69
|
Anton RF, Latham P, Voronin K, Book S, Hoffman M, Prisciandaro J, Bristol E. Efficacy of Gabapentin for the Treatment of Alcohol Use Disorder in Patients With Alcohol Withdrawal Symptoms: A Randomized Clinical Trial. JAMA Intern Med 2020; 180:728-736. [PMID: 32150232 PMCID: PMC7063541 DOI: 10.1001/jamainternmed.2020.0249] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IMPORTANCE Although an estimated 30 million people meet criteria for alcohol use disorder (AUD), few receive appropriate pharmacotherapy. A more personalized, symptom-specific, approach might improve efficacy and acceptance. OBJECTIVE To examine whether gabapentin would be useful in the treatment of AUD, especially in those with the most alcohol withdrawal symptoms. DESIGN, SETTING, AND PARTICIPANTS This double-blind randomized clinical trial conducted between November 2014 and June 2018 evaluated gabapentin vs placebo in community-recruited participants screened and treated in an academic outpatient setting over a 16-week treatment period. A total of 145 treatment-seeking individuals who met Diagnostic and Statistical Manual of Mental Disorders (Fifth Edition) criteria for AUD and were not receiving other AUD intervention were screened, and 96 who also met recent alcohol withdrawal criteria were randomized to treatment after 3 abstinent days. Daily drinking was recorded, and percentage of disialo carbohydrate-deficient transferrin in the blood, a heavy drinking marker, was collected at baseline and monthly during treatment. INTERVENTIONS Gabapentin up to 1200 mg/d, orally, vs placebo along with 9 medical management visits (20 minutes each). MAIN OUTCOMES AND MEASURES The percentage of individuals with no heavy drinking days and those with total abstinence were compared between treatment groups and further evaluated based on prestudy alcohol withdrawal symptoms. RESULTS Of 96 randomized individuals, 90 were evaluable (44 in the gabapentin arm and 46 in the placebo arm), with a mean (SD) age of 49.6 (10.1) years; 69 were men (77%) and 85 were white (94%). The evaluable participants had 83% baseline heavy drinking days (4 or more drinks/day for women, 5 or more for men) and met 4.5 alcohol withdrawal criteria from the Diagnostic and Statistical Manual of Mental Disorders (Fifth Edition). More gabapentin-treated individuals had no heavy drinking days (12 of 44 participants [27%]) compared with placebo (4 of 46 participants [9%]), a difference of 18.6% (95% CI, 3.1-34.1; P = .02; number needed to treat [NNT], 5.4), and more total abstinence (8 of 44 [18%]) compared with placebo (2 of 46 [4%]), a difference of 13.8% (95% CI, 1.0-26.7; P = .04; NNT, 6.2). The prestudy high-alcohol withdrawal group had positive gabapentin effects on no heavy drinking days (P < .02; NNT, 3.1) and total abstinence (P = .003; NNT, 2.7) compared with placebo, while within the low-alcohol withdrawal group, there were no significant differences. These findings were similar for other drinking variables, where gabapentin was more efficacious than placebo in the high-alcohol withdrawal group only. Gabapentin caused more dizziness, but this did not affect efficacy. CONCLUSIONS AND RELEVANCE These data, combined with others, suggest gabapentin might be most efficacious in people with AUD and a history of alcohol withdrawal symptoms. Future studies should evaluate sleep changes and mood during early recovery as mediators of gabapentin efficacy. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02349477.
Collapse
Affiliation(s)
- Raymond F Anton
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - Patricia Latham
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - Konstantin Voronin
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - Sarah Book
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - Michaela Hoffman
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - James Prisciandaro
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| | - Emily Bristol
- Addiction Sciences Division, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston
| |
Collapse
|
70
|
Hammad AM, Sari Y. Effects of Cocaine Exposure on Astrocytic Glutamate Transporters and Relapse-Like Ethanol-Drinking Behavior in Male Alcohol-Preferring Rats. Alcohol Alcohol 2020; 55:254-263. [PMID: 32099993 PMCID: PMC7171926 DOI: 10.1093/alcalc/agaa010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/14/2022] Open
Abstract
AIM Glutamate has been considered as neurotransmitter that is critical in triggering relapse to drugs of abuse, including ethanol and cocaine. Extracellular glutamate concentrations are tightly regulated by several mechanisms, including reuptake through glutamate transporters. Glutamate transporter type 1 (GLT-1) is responsible for clearing the majority of extracellular glutamate. The astrocytic cystine/glutamate antiporter (xCT) regulates also glutamate homeostasis. In this study, we investigated the effects of cocaine exposure and ampicillin/sulbactam (AMP/SUL), a β-lactam antibiotic known to upregulate GLT-1 and xCT, on relapse-like ethanol intake and the expression of astrocytic glutamate transporters in mesocorticolimbic brain regions. METHODS Male alcohol-preferring (P) rats had free access to ethanol for 5 weeks. On Week 6, rats were exposed to either cocaine (20 mg/kg, i.p.) or saline for 12 consecutive days. Ethanol bottles were then removed for 7 days; during the last 5 days, either AMP/SUL (100 or 200 mg/kg, i.p.) or saline was administered to the P rats. Ethanol bottles were reintroduced, and ethanol intake was measured for 4 days. RESULTS Cocaine exposure induced an alcohol deprivation effect (ADE), which was associated in part by a decrease in the expression of GLT-1 and xCT in the nucleus accumbens (NAc) core. AMP/SUL (100 mg/kg, i.p.) attenuated the ADE, while AMP/SUL (200 mg/kg, i.p.) reduced ethanol intake during 4 days of ethanol re-exposure and upregulated GLT-1 and xCT expression in the NAc core, NAc shell and dorsomedial prefrontal cortex (dmPFC). CONCLUSION This study suggests that these astrocytic glutamate transporters might be considered as potential targets for the treatment of polysubstance abuse.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave, Toledo, OH, USA
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130 Amman, 11733, Jordan
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave, Toledo, OH, USA
| |
Collapse
|
71
|
Traumatic brain injury and methamphetamine: A double-hit neurological insult. J Neurol Sci 2020; 411:116711. [DOI: 10.1016/j.jns.2020.116711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/27/2019] [Accepted: 01/29/2020] [Indexed: 11/17/2022]
|
72
|
Back SE, Gray K, Santa Ana E, Jones JL, Jarnecke AM, Joseph JE, Prisciandaro J, Killeen T, Brown DG, Taimina L, Compean E, Malcolm R, Flanagan JC, Kalivas PW. N-acetylcysteine for the treatment of comorbid alcohol use disorder and posttraumatic stress disorder: Design and methodology of a randomized clinical trial. Contemp Clin Trials 2020; 91:105961. [PMID: 32087337 PMCID: PMC7333883 DOI: 10.1016/j.cct.2020.105961] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/06/2020] [Accepted: 02/17/2020] [Indexed: 12/11/2022]
Abstract
Alcohol use disorder (AUD) and posttraumatic stress disorder (PTSD) are two prevalent psychiatric conditions in the U.S. The co-occurrence of AUD and PTSD is also common, and associated with a more severe clinical presentation and worse treatment outcomes across the biopsychosocial spectrum (e.g., social and vocational functioning, physical health) as compared to either disorder alone. Despite the high co-occurrence and negative outcomes, research on effective medications for AUD/PTSD is sparse and there is little empirical evidence to guide treatment decisions. The study described in this paper addresses this knowledge gap by testing the efficacy of N-acetylcysteine (NAC) in reducing alcohol use and PTSD symptoms. Animal studies and prior clinical research suggest a role for NAC in the treatment of substance use disorders and PTSD via glutamate modulation. NAC is a cysteine pro-drug that stimulates the cystine-glutamate exchanger, normalizes glial glutamate transporters, and restores glutamatergic tone on presynaptic receptors in reward regions of the brain. Moreover, NAC is available over-the-counter, has a long-established safety record, and does not require titration to achieve the target dose. This paper describes the rationale, study design, and methodology of a 12-week, randomized, double-blind, placebo-controlled trial of NAC (2400 mg/day) among adults with co-occurring AUD and PTSD. Functional magnetic resonance imaging (fMRI) and proton magnetic resonance spectroscopy (1H-MRS) are utilized to investigate the neural circuitry and neurochemistry underlying comorbid AUD/PTSD and identify predictors of treatment outcome. This study is designed to determine the efficacy of NAC in the treatment of co-occurring AUD/PTSD and provide new information regarding mechanisms of action implicated in co-occurring AUD/PTSD.
Collapse
Affiliation(s)
- Sudie E Back
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Kevin Gray
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Elizabeth Santa Ana
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Jennifer L Jones
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Amber M Jarnecke
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Jane E Joseph
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - James Prisciandaro
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Therese Killeen
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Delisa G Brown
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Linda Taimina
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Ebele Compean
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Robert Malcolm
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Julianne C Flanagan
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Peter W Kalivas
- Department of Neuroscience, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
73
|
Giacometti LL, Barker JM. Sex differences in the glutamate system: Implications for addiction. Neurosci Biobehav Rev 2020; 113:157-168. [PMID: 32173404 DOI: 10.1016/j.neubiorev.2020.03.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 01/21/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022]
Abstract
Clinical and preclinical research have identified sex differences in substance use and addiction-related behaviors. Historically, substance use disorders are more prevalent in men than women, though this gap is closing. Despite this difference, women appear to be more susceptible to the effects of many drugs and progress to substance abuse treatment more quickly than men. While the glutamate system is a key regulator of addiction-related behaviors, much of the work implicating glutamate signaling and glutamatergic circuits has been conducted in men and male rodents. An increasing number of studies have identified sex differences in drug-induced glutamate alterations as well as sex and estrous cycle differences in drug seeking behaviors. This review will describe sex differences in the glutamate system with an emphasis on implications for substance use disorders, highlighting the gaps in our current understanding of how innate and drug-induced alterations in the glutamate system may contribute to sex differences in addiction-related behaviors.
Collapse
Affiliation(s)
- L L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| | - J M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| |
Collapse
|
74
|
Jorge RE, Li R, Liu X, McGavin JK, Shorter DI, Acion L, Arndt S. Treating Alcohol Use Disorder in U.S. Veterans: The Role of Traumatic Brain Injury. J Neuropsychiatry Clin Neurosci 2020; 31:319-327. [PMID: 31117905 DOI: 10.1176/appi.neuropsych.18110250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The authors examined the efficacy of valproate to reduce relapse to heavy drinking among veterans with alcohol use disorder (AUD) and neuropsychiatric comorbidities and whether antecedent traumatic brain injury (TBI) or posttraumatic stress disorder (PTSD) affected treatment response. METHODS Participants were male veterans 18-60 years old with an AUD and no other substance use besides nicotine or cannabis. Sixty-two patients were randomly assigned to receive either valproate or naltrexone. Participants were evaluated at baseline and followed weekly for 24 weeks. All participants received standardized psychosocial interventions as well as treatment for coexistent psychiatric conditions. RESULTS During the follow-up period, nine study subjects in the naltrexone group and 14 in the valproate group relapsed to heavy drinking, but the difference did not reach statistical significance. Participants with a history of moderate to severe TBI were more likely to relapse to heavy drinking compared with those with no TBI (hazard ratio=4.834, 95% CI=1.103-21.194, p=0.033). PTSD status did not significantly affect outcome. CONCLUSIONS Intensive outpatient programs are efficacious alternatives to treat AUD in veterans, although the role of pharmacological treatment is not completely elucidated. Glutamatergic agents appear to be less effective than opiate antagonists to prevent relapse to heavy drinking and to increase cumulative abstinence. Future studies should examine novel pharmacological and nonpharmacological options.
Collapse
Affiliation(s)
- Ricardo E Jorge
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Ruosha Li
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Xiangyu Liu
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Jill K McGavin
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Daryl I Shorter
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Laura Acion
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| | - Stephan Arndt
- The Mental Health Care Line, Michael E. DeBakey VA Medical Center, Houston (Jorge, McGavin, Shorter, Acion); the Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Jorge, McGavin, Shorter, Acion); the Department of Biostatistics and Data Science, School of Public Health, University of Texas Health Science Center at Houston (Li, Liu); the Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City (Acion, Arndt); and the Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City (Arndt)
| |
Collapse
|
75
|
Influence of combined treatment with naltrexone and memantine on alcohol drinking behaviors: a phase II randomized crossover trial. Neuropsychopharmacology 2020; 45:319-326. [PMID: 31590179 PMCID: PMC6901445 DOI: 10.1038/s41386-019-0536-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/17/2022]
Abstract
Glutamate and opioid systems play important roles in alcohol drinking behaviors. We examined if combined treatment with the NMDA antagonist memantine and the opioid antagonist naltrexone, when compared with naltrexone alone, would have a greater influence on alcohol drinking behaviors. Fifty-six, non-treatment-seeking heavy drinkers, with alcohol dependence and a positive family history (FHP) of alcoholism, participated in a randomized, double-blind, crossover trial, including two 6-8 days treatment periods, separated by a 6-day washout, and 3 alcohol drinking paradigm (ADP) sessions. After the first baseline (BAS) ADP1 session, participants were randomized to receive either naltrexone (NTX; 50 mg/day) + placebo memantine, or NTX (50 mg/day) + memantine (MEM; 20 mg/day), during the first treatment period, following which they completed ADP2. After a 6-day washout, participants were crossed over to the treatment they did not receive during the first treatment period, following which they completed ADP3. During each ADP, participants received a priming drink of alcohol followed by 3 1-hour, self-administration periods during which they had ad-lib access to 12 drinks. Individually, both NTX and NTX + MEM, when compared to BAS ADP1, significantly reduced the number of drinks consumed (p's < 0.001) and craving (p's < 0.001). When comparing NTX + MEM vs. NTX on number of drinks consumed, there was a significant treatment* sequence interaction (p = 0.004). Specifically, when NTX + MEM followed NTX alone, NTX + MEM resulted in a further reduction in drinking (mean: -1.94; 95% CI: -2.6, -0.8, p = 0.0005). However, when NTX alone followed NTX + MEM, NTX alone did not lead to further reduction in drinking (mean: 0.59; 95% CI: -0.67, 1.43, p = 0.47). Similar patterns were observed for alcohol craving; specifically, a significant reduction in craving was observed when NTX + MEM followed NTX alone (p = 0.009), but craving reduction was maintained when NTX + MEM was followed by NTX alone. Neither treatment condition significantly influenced alcohol-induced stimulation or sedation. Memantine (at a dose of 20 mg/day) enhances the efficacy of naltrexone (50 mg/day) in reducing alcohol drinking and craving among FHP drinkers with beneficial effects that appear to carryover after discontinuation of memantine treatment.
Collapse
|
76
|
Chen J, Liu Q, Fan R, Han H, Yang Z, Cui W, Song G, Li MD. Demonstration of critical role of GRIN3A in nicotine dependence through both genetic association and molecular functional studies. Addict Biol 2020; 25:e12718. [PMID: 30741440 DOI: 10.1111/adb.12718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/19/2018] [Accepted: 01/08/2019] [Indexed: 11/27/2022]
Abstract
Nicotine dependence (ND) is a chronic disease with catastrophic effects on individual and public health. The glutamate receptor subunit gene, ionotropic N-methyl-d-aspartate 3A (GRIN3A), encodes a crucial subunit of N-methyl-d-aspartate receptors (NMDARs), which play an essential role in synaptic plasticity in the brain. Although various variants of GRIN3A have been associated with ND in European-American and African-American samples, no study has been reported for the association between GRIN3A and ND in Chinese Han population. We performed an association study of 16 single nucleotide polymorphisms (SNPs) in GRIN3A with ND in 2616 Chinese individuals. SNP-based association analysis indicated that SNP rs1323423 was significantly associated with the Fagerström Test for Nicotine Dependence (FTND) score after correction for multiple testing (P = 0.0026). Haplotype-based association analysis revealed that Block 3, formed by rs1323423-rs10989591, was significantly associated with the FTND score after correction for multiple testing (global P = 0.0183). Furthermore, luciferase reporter assay demonstrated that the DNA region containing rs1323423 was an enhancer element, the activity of which was significantly impacted by rs1323423 genotype. Considering that rs1323423 is located in a potential enhancer region, we performed GRIN3A editing in HEK293T cells with CRISPR/Cas9 and found that the DNA region around rs1323423 has a regulatory function and the expression of GRIN3A affects the expression of other NMDA subunits. Moreover, we demonstrated that nicotine at a concentration of 100 μM decreased expression of GRIN3A in SH-SY5Y and HEK293T cells at the RNA and protein level, respectively. This study provides novel evidence for the involvement of GRIN3A in ND.
Collapse
Affiliation(s)
- Jiali Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Qiang Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Rongli Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Haijun Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Wenyan Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
| | - Guohua Song
- Animal Research CenterShanxi Medical University China
| | - Ming D. Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesZhejiang University School of Medicine China
- Research Center for Air Pollution and HealthZhejiang University China
- Institute of NeuroImmune PharmacologySeton Hall University South Orange New Jersey USA
| |
Collapse
|
77
|
Karthivashan G, Ganesan P, Park SY, Lee HW, Choi DK. Lipid-based nanodelivery approaches for dopamine-replacement therapies in Parkinson's disease: From preclinical to translational studies. Biomaterials 2019; 232:119704. [PMID: 31901690 DOI: 10.1016/j.biomaterials.2019.119704] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022]
Abstract
The incidence of Parkinson's disease (PD), the second most common neurodegenerative disorder, has increased exponentially as the global population continues to age. Although the etiological factors contributing to PD remain uncertain, its average incidence rate is reported to be 1% of the global population older than 60 years. PD is primarily characterized by the progressive loss of dopaminergic (DAergic) neurons and/or associated neuronal networks and the subsequent depletion of dopamine (DA) levels in the brain. Thus, DA or levodopa (l-dopa), a precursor of DA, represent cardinal targets for both idiopathic and symptomatic PD therapeutics. While several therapeutic strategies have been investigated over the past decade for their abilities to curb the progression of PD, an effective cure for PD is currently unavailable. Even DA replacement therapy, an effective PD therapeutic strategy that provides an exogenous supply of DA or l-dopa, has been hindered by severe challenges, such as a poor capacity to bypass the blood-brain barrier and inadequate bioavailability. Nevertheless, with recent advances in nanotechnology, several drug delivery systems have been developed to bypass the barriers associated with central nervous system therapeutics. In here, we sought to describe the adapted lipid-based nanodrug delivery systems used in the field of PD therapeutics and their recent advances, with a particular focus placed on DA replacement therapies. This work initially explores the background of PD; offers descriptions of the most recent molecular targets; currently available clinical medications/limitations; an overview of several lipid-based PD nanotherapeutics, functionalized nanoparticles, and technical aspects in brain delivery; and, finally, presents future perspectives to enhance the use of nanotherapeutics in PD treatment.
Collapse
Affiliation(s)
- Govindarajan Karthivashan
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea; Research Institute of Inflammatory Diseases (RID), College of Biomedical and Health Science and BK21plus Glocal Education Program of Nutraceuticals Development, Konkuk University, Chungju, 27478, Republic of Korea
| | - Palanivel Ganesan
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea; Department of Biomedical Chemistry, Nanotechnology Research Center, Department of Applied Life Science, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea
| | - Shin-Young Park
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Kyungpook National University School of Medicine and Brain Science & Engineering Institute, Kyungpook National University, Daegu, 41404, Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea; Research Institute of Inflammatory Diseases (RID), College of Biomedical and Health Science and BK21plus Glocal Education Program of Nutraceuticals Development, Konkuk University, Chungju, 27478, Republic of Korea.
| |
Collapse
|
78
|
Arezoomandan R, Aliaghaei A, Khodagholi F, Haghparast A. Minocycline induces the expression of intra-accumbal glutamate transporter-1 in the morphine-dependent rats. Asian J Psychiatr 2019; 46:70-73. [PMID: 31630007 DOI: 10.1016/j.ajp.2019.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
Glial glutamate transporters (GLT-1) is responsible for glutamate homeostasis. GLT-1 expression and glutamate uptake can be affected by addictive drugs and can be used as a target in addiction pharmacotherapy. It has been shown that minocycline, an antibiotic with anti-inflammatory, and neuroprotective properties, can upregulate the expression of GLT-1. In the present study, in morphine-dependent rats, the effect of minocycline on expression of GLT-1 in nucleus accumbens was investigated by immunohistochemistry. The expression of GLT-1 significantly increased in minocycline treated animals. In line with other studies, our findings showed that restoring GLT-1 expression with minocycline might be considered as a potential target for correcting pre-clinical and clinical manifestations of drug addiction.
Collapse
Affiliation(s)
- Reza Arezoomandan
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Neuroscience Lab, Biology and Anatomical Sciences Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
79
|
Nunes PT, Kipp BT, Reitz NL, Savage LM. Aging with alcohol-related brain damage: Critical brain circuits associated with cognitive dysfunction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:101-168. [PMID: 31733663 PMCID: PMC7372724 DOI: 10.1016/bs.irn.2019.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcoholism is associated with brain damage and impaired cognitive functioning. The relative contributions of different etiological factors, such as alcohol, thiamine deficiency and age vulnerability, to the development of alcohol-related neuropathology and cognitive impairment are still poorly understood. One reason for this quandary is that both alcohol toxicity and thiamine deficiency produce brain damage and cognitive problems that can be modulated by age at exposure, aging following alcohol toxicity or thiamine deficiency, and aging during chronic alcohol exposure. Pre-clinical models of alcohol-related brain damage (ARBD) have elucidated some of the contributions of ethanol toxicity and thiamine deficiency to neuroinflammation, neuronal loss and functional deficits. However, the critical variable of age at the time of exposure or long-term aging with ARBD has been relatively ignored. Acute thiamine deficiency created a massive increase in neuroimmune genes and proteins within the thalamus and significant increases within the hippocampus and frontal cortex. Chronic ethanol treatment throughout adulthood produced very minor fluctuations in neuroimmune genes, regardless of brain region. Intermittent "binge-type" ethanol during the adolescent period established an intermediate neuroinflammatory response in the hippocampus and frontal cortex, that can persist into adulthood. Chronic excessive drinking throughout adulthood, adolescent intermittent ethanol exposure, and thiamine deficiency all led to a loss of the cholinergic neuronal phenotype within the basal forebrain, reduced hippocampal neurogenesis, and alterations in the frontal cortex. Only thiamine deficiency results in gross pathological lesions of the thalamus. The behavioral impairment following these types of treatments is hierarchical: Thiamine deficiency produces the greatest impairment of hippocampal- and prefrontal-dependent behaviors, chronic ethanol drinking ensues mild impairments on both types of tasks and adolescent intermittent ethanol exposure leads to impairments on frontocortical tasks, with sparing on most hippocampal-dependent tasks. However, our preliminary data suggest that as rodents age following adolescent intermittent ethanol exposure, hippocampal functional deficits began to emerge. A necessary requirement for the advancement of understanding the neural consequences of alcoholism is a more comprehensive assessment and understanding of how excessive alcohol drinking at different development periods (adolescence, early adulthood, middle-aged and aged) influences the trajectory of the aging process, including pathological aging and disease.
Collapse
Affiliation(s)
- Polliana Toledo Nunes
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Brian T Kipp
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Nicole L Reitz
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| | - Lisa M Savage
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States.
| |
Collapse
|
80
|
Bamford NS, Wang W. Corticostriatal plasticity in the nucleus accumbens core. J Neurosci Res 2019; 97:1559-1578. [PMID: 31298422 DOI: 10.1002/jnr.24494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 11/06/2022]
Abstract
Small fluctuations in striatal glutamate and dopamine are required to establish goal-directed behaviors and motor learning, while large changes appear to underlie many neuropsychological disorders, including drug dependence and Parkinson's disease. A better understanding of how variations in neurotransmitter availability can modify striatal circuitry will lead to new therapeutic targets for these disorders. Here, we examined dopamine-induced plasticity in prefrontal cortical projections to the nucleus accumbens (NAc) core. We combined behavioral measures of male mice, presynaptic optical studies of glutamate release kinetics from prefrontal cortical projections, and postsynaptic electrophysiological recordings of spiny projection neurons within the NAc core. Our data show that repeated amphetamine promotes long-lasting but reversible changes along the corticoaccumbal pathway. In saline-treated mice, coincident cortical stimulation and dopamine release promoted presynaptic filtering by depressing exocytosis from glutamatergic boutons with a low-probability of release. The repeated use of amphetamine caused a frequency-dependent, progressive, and long-lasting depression in corticoaccumbal activity during withdrawal. This chronic presynaptic depression was relieved by a drug challenge which potentiated glutamate release from synapses with a low-probability of release. D1 receptors generated this synaptic potentiation, which corresponded with the degree of locomotor sensitization in individual mice. By reversing the synaptic depression, drug reinstatement may promote allostasis by returning corticoaccumbal activity to a more stable and normalized state. Therefore, dopamine-induced synaptic filtering of excitatory signals entering the NAc core in novice mice and paradoxical excitation of the corticoaccumbal pathway during drug reinstatement may encode motor learning, habit formation, and dependence.
Collapse
Affiliation(s)
- Nigel S Bamford
- Department of Pediatrics, Yale University, New Haven, Connecticut.,Department of Neurology, Yale University, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut.,Department of Neurology, University of Washington, Seattle, Washington
| | - Wengang Wang
- Department of Neurology, University of Washington, Seattle, Washington.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
81
|
Lainiola M, Hietala L, Linden AM, Aitta-Aho T. The lack of conditioned place preference, but unaltered stimulatory and ataxic effects of alcohol in mGluR3-KO mice. J Psychopharmacol 2019; 33:855-864. [PMID: 31070489 DOI: 10.1177/0269881119844178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Alcohol use associates with environmental cues that can later reinstate drinking patterns without any alcohol exposure. Alcohol-induced reward, when combined with contextual signals of various sensory modalities in the central synapses of mesolimbic reward circuitries, can lead to the formation of conditioned responses. AIMS As the activation of glutamatergic synapses is pivotal in such processes, we aimed to investigate whether the metabotropic glutamate receptor subtype 3 plays a role in alcohol-induced behaviours including place preference. METHODS The metabotropic glutamate receptor subtype 3 knockout (mGluR3-KO) mouse line was used to study alcohol-induced place preference, locomotor activating and ataxic effects, limited access alcohol drinking, and preference for sucrose and saccharin. RESULTS Alcohol-induced horizontal locomotor stimulation and reduced rearing behaviour remained unchanged in the mGluR3-KO mice. However, alcohol-induced place conditioning in an unbiased paradigm setup was lacking in the mGluR3-KO mice, but clearly present in wildtype mice. Locomotor activity was not different between the mGluR3-KO and wildtype mice during the acquisition and expression trials. Alcohol consumption, studied through the 'drinking in the dark' model, remained unchanged in the mGluR3-KO mice, although low consumption in both wildtype and knockout mice hampers interpretation. The mGluR3-KO mice also showed normal sucrose and saccharin preference. CONCLUSIONS These studies indicate a role for metabotropic glutamate receptor subtype 3 in the conditioned contextual alcohol responses, but not in stimulatory, and ataxic alcohol effects.
Collapse
Affiliation(s)
- Mira Lainiola
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Lana Hietala
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anni-Maija Linden
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
82
|
Bolewska P, Martin BI, Orlando KA, Rhoads DE. Sequential Changes in Brain Glutamate and Adenosine A1 Receptors May Explain Severity of Adolescent Alcohol Withdrawal after Consumption of High Levels of Alcohol. NEUROSCIENCE JOURNAL 2019; 2019:5950818. [PMID: 31275953 PMCID: PMC6582803 DOI: 10.1155/2019/5950818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/28/2019] [Indexed: 01/06/2023]
Abstract
There is an excellent correlation between the age when alcohol consumption begins and the likelihood of lifelong problems with alcohol abuse. Alcohol use often begins in adolescence, a time marked by brain development and maturation of numerous brain systems. Rats are an important model, wherein the emergence of alcohol withdrawal symptoms serves as a gauge of dependency following chronic alcohol consumption. Previous work has shown that adolescent Long-Evans rats consume high levels of alcohol and develop a severe alcohol withdrawal syndrome when fed alcohol as part of a liquid diet. Acutely, alcohol inhibits two important excitatory receptors for glutamate (NMDA and AMPA) and may further decrease glutamate activity through modulatory adenosine receptors. The present study focuses on potential adaptive changes in expression of these receptors that may create a receptor imbalance during chronic alcohol consumption and lead to severe overexcitation of the adolescent brain during alcohol withdrawal. Levels of brain expression of NMDA, AMPA, and adenosine A1 and A2a receptors were determined by Western blotting after adolescent rats consumed an alcohol-containing liquid diet for 4, 11, or 18 days. Severity of alcohol withdrawal was also assessed at these time points. Levels increased for both AMPA and NMDA receptors, significant and approaching maximal by day 11. In contrast, A1 receptor density showed a slow decline reaching significance at 18 days. There were no changes in expression of adenosine A2a receptor. The most severe withdrawal symptoms appear to coincide with the later downregulation of adenosine A1 receptors coming on top of maximal upregulation of excitatory AMPA and NMDA glutamate receptors. Thus, loss of adenosine "brakes" on glutamate excitation may punctuate receptor imbalance in alcohol-consuming adolescents by allowing the upregulation of the excitatory receptors to have full impact during early alcohol withdrawal.
Collapse
Affiliation(s)
- Patrycja Bolewska
- Department of Biology, Monmouth University, W. Long Branch, NJ 07764, USA
| | - Bryan I. Martin
- Department of Biology, Monmouth University, W. Long Branch, NJ 07764, USA
| | - Krystal A. Orlando
- Department of Biology, Monmouth University, W. Long Branch, NJ 07764, USA
| | - Dennis E. Rhoads
- Department of Biology, Monmouth University, W. Long Branch, NJ 07764, USA
| |
Collapse
|
83
|
Meda SA, Narayanan B, Chorlian D, Meyers JL, Gelernter J, Hesselbrock V, Bauer L, Calhoun VD, Porjesz B, Pearlson GD. Multivariate Analyses Reveal Biological Components Related to Neuronal Signaling and Immunity Mediating Electroencephalograms Abnormalities in Alcohol-Dependent Individuals from the Collaborative Study on the Genetics of Alcoholism Cohort. Alcohol Clin Exp Res 2019; 43:1462-1477. [PMID: 31009096 DOI: 10.1111/acer.14063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The underlying molecular mechanisms associated with alcohol use disorder (AUD) risk have only been partially revealed using traditional approaches such as univariate genomewide association and linkage-based analyses. We therefore aimed to identify gene clusters related to Electroencephalograms (EEG) neurobiological phenotypes distinctive to individuals with AUD using a multivariate approach. METHODS The current project adopted a bimultivariate data-driven approach, parallel independent component analysis (para-ICA), to derive and explore significant genotype-phenotype associations in a case-control subset of the Collaborative Study on the Genetics of Alcoholism (COGA) dataset. Para-ICA subjects comprised N = 799 self-reported European Americans (367 controls and 432 AUD cases), recruited from COGA, who had undergone resting EEG and genotyping. Both EEG and genomewide single nucleotide polymorphism (SNP) data were preprocessed prior to being subjected to para-ICA in order to derive genotype-phenotype relationships. RESULTS From the data, 4 EEG frequency and 4 SNP components were estimated, with 2 significantly correlated EEG-genetic relationship pairs. The first such pair primarily represented theta activity, negatively correlated with a genetic cluster enriched for (but not limited to) ontologies/disease processes representing cell signaling, neurogenesis, transmembrane drug transportation, alcoholism, and lipid/cholesterol metabolism. The second component pair represented mainly alpha activity, positively correlated with a genetic cluster with ontologies similarly enriched as the first component. Disease-related enrichments for this component revealed heart and autoimmune disorders as top hits. Loading coefficients for both the alpha and theta components were significantly reduced in cases compared to controls. CONCLUSIONS Our data suggest plausible multifactorial genetic components, primarily enriched for neuronal/synaptic signaling/transmission, immunity, and neurogenesis, mediating low-frequency alpha and theta abnormalities in alcohol addiction.
Collapse
Affiliation(s)
- Shashwath A Meda
- Olin Neuropsychiatry Research Center, Hartford Hospital/IOL, Hartford, Connecticut
| | - Balaji Narayanan
- Olin Neuropsychiatry Research Center, Hartford Hospital/IOL, Hartford, Connecticut
| | - David Chorlian
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, New York
| | - Jacquelyn L Meyers
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, New York
| | - Joel Gelernter
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| | | | - Lance Bauer
- Department of Psychiatry, UConn Health, Farmington, Connecticut
| | | | - Bernice Porjesz
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, New York
| | - Godfrey D Pearlson
- Olin Neuropsychiatry Research Center, Hartford Hospital/IOL, Hartford, Connecticut.,Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
84
|
Wen X, Wang B, Huang S, Liu TL, Lee MS, Chung PS, Chow YT, Huang IW, Monbouquette HG, Maidment NT, Chiou PY. Flexible, multifunctional neural probe with liquid metal enabled, ultra-large tunable stiffness for deep-brain chemical sensing and agent delivery. Biosens Bioelectron 2019; 131:37-45. [PMID: 30818131 PMCID: PMC6602555 DOI: 10.1016/j.bios.2019.01.060] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 10/27/2022]
Abstract
Flexible neural probes have been pursued previously to minimize the mechanical mismatch between soft neural tissues and implants and thereby improve long-term performance. However, difficulties with insertion of such probes deep into the brain severely restricts their utility. We describe a solution to this problem using gallium (Ga) in probe construction, taking advantage of the solid-to-liquid phase change of the metal at body temperature and probe shape deformation to provide temperature-dependent control of stiffness over 5 orders of magnitude. Probes in the stiff state were successfully inserted 2 cm-deep into agarose gel "brain phantoms" and into rat brains under cooled conditions where, upon Ga melting, they became ultra soft, flexible, and stretchable in all directions. The current 30 μm-thick probes incorporated multilayer, deformable microfluidic channels for chemical agent delivery, electrical interconnects through Ga wires, and high-performance electrochemical glutamate sensing. These PDMS-based microprobes of ultra-large tunable stiffness (ULTS) should serve as an attractive platform for multifunctional chronic neural implants.
Collapse
Affiliation(s)
- Ximiao Wen
- Department of Mechanical and Aerospace Engineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - Bo Wang
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, CA, USA
| | - Shan Huang
- Department of Biological Chemistry, University of California at Los Angeles, CA, USA
| | - Tingyi Leo Liu
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, MA, USA
| | - Meng-Shiue Lee
- Department of Mechanical Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Shan Chung
- Department of Bioengineering, University of California at Los Angeles, CA, USA
| | - Yu Ting Chow
- Department of Mechanical and Aerospace Engineering, University of California at Los Angeles, Los Angeles, CA, USA
| | - I-Wen Huang
- Department of Chemical and Biomolecular Engineering, University of California at Los Angeles, CA, USA
| | - Harold G Monbouquette
- Department of Chemical and Biomolecular Engineering, University of California at Los Angeles, CA, USA
| | - Nigel T Maidment
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, CA, USA.
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering, University of California at Los Angeles, Los Angeles, CA, USA; Department of Bioengineering, University of California at Los Angeles, CA, USA.
| |
Collapse
|
85
|
Prabhu VV, Nguyen TB, Cui Y, Oh YE, Piao YH, Baek HM, Kim JY, Shin KH, Kim JH, Lee KH, Chung YC. Metabolite signature associated with stress susceptibility in socially defeated mice. Brain Res 2019; 1708:171-180. [DOI: 10.1016/j.brainres.2018.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 11/13/2018] [Accepted: 12/15/2018] [Indexed: 01/10/2023]
|
86
|
Ikonne US, Vann PH, Wong JM, Forster MJ, Sumien N. Supplementation with N-Acetyl Cysteine Affects Motor and Cognitive Function in Young but Not Old Mice. J Nutr 2019; 149:463-470. [PMID: 30770531 PMCID: PMC6398433 DOI: 10.1093/jn/nxy287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/14/2018] [Accepted: 10/18/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND N-acetyl cysteine (NAC) is a thiolic antioxidant that is thought to increase cellular glutathione (GSH) by augmenting the concentration of available cysteine, an essential precursor to GSH production. Manipulating redox status can affect brain function, and NAC intake has been associated with improving brain function in models of neurodegenerative diseases. OBJECTIVES The objective of the study was to determine if short-term dietary supplementation with NAC could ameliorate functional impairment associated with aging. METHODS C57BL/6J male mice aged 6, 12, or 24 mo were fed a control diet or the control diet supplemented with 0.3% NAC for a total of 12 wk. After 4 wk of dietary supplementation, mice began a series of behavioral tests to measure spontaneous activity (locomotor activity test), psychomotor performance (bridge-walking and coordinated running), and cognitive capacity (Morris water maze and discriminated active avoidance). The performance of the mice on these tests was analyzed through the use of analyses of variance with Age and Diet as factors. RESULTS Supplementation of NAC improved peak motor performance in a coordinated running task by 14% (P < 0.05), and increased the time spent around the platform by 24% in a Morris water maze at age 6 mo. However, the supplementation had no to minimal effect on the motor and cognitive functions of 12- and 24-mo-old mice. CONCLUSIONS The findings of this preclinical study support the claim that NAC has nootropic properties in 6-mo-old mice, but suggest that it may not be useful for improving motor and cognitive impairments in older mice.
Collapse
Affiliation(s)
- Uzoma S Ikonne
- Department of Pharmacology and Neuroscience and Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX,Basic Medical Science, School of Osteopathic Medicine Arizona, A.T. Still University, Mesa, AZ
| | - Philip H Vann
- Department of Pharmacology and Neuroscience and Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX
| | - Jessica M Wong
- Department of Pharmacology and Neuroscience and Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX
| | - Michael J Forster
- Department of Pharmacology and Neuroscience and Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience and Institute for Healthy Aging, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX,Address correspondence to NS (e-mail: )
| |
Collapse
|
87
|
Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, Datta G, Davila-Velderrain J, McGuire D, Tian C, Zhan X, Choquet H, Docherty AR, Faul JD, Foerster JR, Fritsche LG, Gabrielsen ME, Gordon SD, Haessler J, Hottenga JJ, Huang H, Jang SK, Jansen PR, Ling Y, Mägi R, Matoba N, McMahon G, Mulas A, Orrù V, Palviainen T, Pandit A, Reginsson GW, Skogholt AH, Smith JA, Taylor AE, Turman C, Willemsen G, Young H, Young KA, Zajac GJM, Zhao W, Zhou W, Bjornsdottir G, Boardman JD, Boehnke M, Boomsma DI, Chen C, Cucca F, Davies GE, Eaton CB, Ehringer MA, Esko T, Fiorillo E, Gillespie NA, Gudbjartsson DF, Haller T, Harris KM, Heath AC, Hewitt JK, Hickie IB, Hokanson JE, Hopfer CJ, Hunter DJ, Iacono WG, Johnson EO, Kamatani Y, Kardia SLR, Keller MC, Kellis M, Kooperberg C, Kraft P, Krauter KS, Laakso M, Lind PA, Loukola A, Lutz SM, Madden PAF, Martin NG, McGue M, McQueen MB, Medland SE, Metspalu A, Mohlke KL, Nielsen JB, Okada Y, Peters U, Polderman TJC, Posthuma D, Reiner AP, Rice JP, Rimm E, Rose RJ, Runarsdottir V, Stallings MC, Stančáková A, Stefansson H, Thai KK, Tindle HA, Tyrfingsson T, Wall TL, Weir DR, Weisner C, Whitfield JB, Winsvold BS, Yin J, Zuccolo L, Bierut LJ, Hveem K, Lee JJ, Munafò MR, Saccone NL, Willer CJ, Cornelis MC, David SP, Hinds DA, Jorgenson E, Kaprio J, Stitzel JA, Stefansson K, Thorgeirsson TE, Abecasis G, Liu DJ, Vrieze S. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat Genet 2019; 51:237-244. [PMID: 30643251 PMCID: PMC6358542 DOI: 10.1038/s41588-018-0307-5] [Citation(s) in RCA: 1141] [Impact Index Per Article: 228.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 11/06/2018] [Indexed: 12/21/2022]
Abstract
Tobacco and alcohol use are leading causes of mortality that influence risk for many complex diseases and disorders1. They are heritable2,3 and etiologically related4,5 behaviors that have been resistant to gene discovery efforts6-11. In sample sizes up to 1.2 million individuals, we discovered 566 genetic variants in 406 loci associated with multiple stages of tobacco use (initiation, cessation, and heaviness) as well as alcohol use, with 150 loci evidencing pleiotropic association. Smoking phenotypes were positively genetically correlated with many health conditions, whereas alcohol use was negatively correlated with these conditions, such that increased genetic risk for alcohol use is associated with lower disease risk. We report evidence for the involvement of many systems in tobacco and alcohol use, including genes involved in nicotinic, dopaminergic, and glutamatergic neurotransmission. The results provide a solid starting point to evaluate the effects of these loci in model organisms and more precise substance use measures.
Collapse
Affiliation(s)
- Mengzhen Liu
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Yu Jiang
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Robbee Wedow
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Yue Li
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David M Brazel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Interdisciplinary Quantitative Biology Graduate Group, University of Colorado Boulder, Boulder, CO, USA
| | - Fang Chen
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Gargi Datta
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Jose Davila-Velderrain
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel McGuire
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Chao Tian
- 23andMe, Inc., Mountain View, CA, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for the Genetics of Host Defense, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Anna R Docherty
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry and Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Johanna R Foerster
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Lars G Fritsche
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Maiken Elvestad Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Scott D Gordon
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hongyan Huang
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Seon-Kyeong Jang
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Philip R Jansen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Yueh Ling
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Nana Matoba
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - George McMahon
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Anita Pandit
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Amy E Taylor
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hannah Young
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Kendra A Young
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gregory J M Zajac
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | | | - Jason D Boardman
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Chu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | | | - Charles B Eaton
- Department of Family Medicine and Community Health, Alpert Medical School, Brown University, Providence, RI, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Tõnu Esko
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Nathan A Gillespie
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew C Heath
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - John K Hewitt
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - John E Hokanson
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian J Hopfer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - William G Iacono
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Eric O Johnson
- Fellows Program, RTI International, Research Triangle Park, NC, USA
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kenneth S Krauter
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Markku Laakso
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anu Loukola
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sharon M Lutz
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Pamela A F Madden
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Matt McGue
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Matthew B McQueen
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonas B Nielsen
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Tinca J C Polderman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Genetics, VU Medical Centre Amsterdam, Amsterdam, the Netherlands
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - John P Rice
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Michael C Stallings
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Alena Stančáková
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | | | - Khanh K Thai
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hilary A Tindle
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | | | - Tamara L Wall
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Constance Weisner
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Jie Yin
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Luisa Zuccolo
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - James J Lee
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Psychological Science, University of Bristol, Bristol, UK
| | - Nancy L Saccone
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Marilyn C Cornelis
- Department of Preventative Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sean P David
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Gonçalo Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Dajiang J Liu
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA.
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA.
| | - Scott Vrieze
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
88
|
Cannizzaro C, Talani G, Brancato A, Mulas G, Spiga S, De Luca MA, Sanna A, Marino RAM, Biggio G, Sanna E, Diana M. Dopamine Restores Limbic Memory Loss, Dendritic Spine Structure, and NMDAR-Dependent LTD in the Nucleus Accumbens of Alcohol-Withdrawn Rats. J Neurosci 2019; 39:929-943. [PMID: 30446531 PMCID: PMC6382989 DOI: 10.1523/jneurosci.1377-18.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023] Open
Abstract
Alcohol abuse leads to aberrant forms of emotionally salient memory, i.e., limbic memory, that promote escalated alcohol consumption and relapse. Accordingly, activity-dependent structural abnormalities are likely to contribute to synaptic dysfunctions that occur from suddenly ceasing chronic alcohol consumption. Here we show that alcohol-dependent male rats fail to perform an emotional-learning task during abstinence but recover their functioning by l-3,4-dihydroxyphenylalanin (l-DOPA) administration during early withdrawal. l-DOPA also reverses the selective loss of dendritic "long thin" spines observed in medium spiny neurons of the nucleus accumbens (NAc) shell of alcohol-dependent rats during abstinence, as well as the reduction in tyrosine hydroxylase immunostaining and postsynaptic density-95-positive elements. Patch-clamp experiments in NAc slices reveal that both in vivo systemic l-DOPA administration and in vitro exposure to dopamine can restore the loss of long-term depression (LTD) formation, counteract the reduction in NMDAR-mediated synaptic currents and rectify the altered NMDAR/AMPAR ratio observed in alcohol-withdrawn rats. Further, in vivo microdialysis experiments show that blunted dopaminergic signaling is revived after l-DOPA treatment during early withdrawal. These results suggest a key role of an efficient dopamine signaling for maintaining, and restore, neural trophism, NMDA-dependent LTD, and ultimately optimal learning.SIGNIFICANCE STATEMENT Blunted dopamine signaling and altered glutamate connectivity in the nucleus accumbens represent the neuroanatomical basis for the impairment in aversive limbic memory observed during withdrawal in alcohol dependence. Supplying l-DOPA during withdrawal re-establishes synaptic morphology and functional neuroadaptations, suggesting a complete recovery of nucleus accumbens glutamatergic synaptic plasticity when dopamine is revived. Importantly, restoring dopamine transmission allows those synapses to encode emotionally relevant information and rescue flexibility in the neuronal circuits that process limbic memory formation. Under these conditions, drugs capable of selectively boosting the dopaminergic function during the "fluid" and still responsive state of the early withdrawn maladaptive synapses may help in the treatment of alcohol addiction.
Collapse
Affiliation(s)
- Carla Cannizzaro
- Laboratory of Neuropsychopharmacology, Department ProSaMI G. D'Alessandro, University of Palermo, Via del Vespro 129 90127, Palermo, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
| | - Anna Brancato
- Laboratory of Neuropsychopharmacology, Department ProSaMI G. D'Alessandro, University of Palermo, Via del Vespro 129 90127, Palermo, Italy
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cittadella Universitaria-S.P., 09042 Monserrato, Cagliari, Italy
| | - Angela Sanna
- Department of Medical Science and Public Health, University of Cagliari, Cittadella Universitaria-S.P., 09042 Monserrato, Cagliari, Italy
| | - Rosa Anna Maria Marino
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, and
| | - Giovanni Biggio
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Enrico Sanna
- Institute of Neuroscience, National Research Council, 09042 Monserrato, Cagliari, Italy
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Marco Diana
- University of Sassari, G.Minardi Laboratory of Cognitive Neuroscience, Department of Chemistry and Pharmacy Via Muroni, 23 07100 Sassari, Italy
| |
Collapse
|
89
|
Gerace E, Landucci E, Bani D, Moroni F, Mannaioni G, Pellegrini-Giampietro DE. Glutamate Receptor-Mediated Neurotoxicity in a Model of Ethanol Dependence and Withdrawal in Rat Organotypic Hippocampal Slice Cultures. Front Neurosci 2019; 12:1053. [PMID: 30733663 PMCID: PMC6353783 DOI: 10.3389/fnins.2018.01053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/27/2018] [Indexed: 11/15/2022] Open
Abstract
Long-term alcohol use can lead to alterations in brain structure and functions and, in some cases, to neurodegeneration. Several mechanisms have been proposed to explain ethanol (EtOH)-related brain injury. One of the most relevant mechanisms of alcohol-induced neurodegeneration involves glutamatergic transmission, but their exact role is not yet fully understood. We investigated the neurochemical mechanisms underlying the toxicity induced by EtOH dependence and/or withdrawal by exposing rat organotypic hippocampal slices to EtOH (100–300 mM) for 7 days and then incubating the slices in EtOH-free medium for the subsequent 24 h. EtOH withdrawal led to a dose-dependent CA1 pyramidal cell injury, as detected with propidium iodide fluorescence. Electron microscopy of hippocampal slices revealed that not only EtOH withdrawal but also 7 days chronic EtOH exposure elicited signs of apoptotic cell death in CA1 pyramidal cells. These data were supported by electrophysiological recordings of spontaneus Excitatory Post Synaptic Currents (sEPSCs) from CA1 pyramidal cells. The average amplitude of sEPSCs in slices treated with EtOH for 7 days was significantly increased, and even more so during the first 30 min of EtOH withdrawal, suggesting that the initial phase of the neurodegenerative process could be due to an excitotoxic mechanism. We then analyzed the expression levels of presynaptic (vGlut1, vGlut2, CB1 receptor, synaptophysin) and postsynaptic (PSD95, GluN1, GluN2A, GluN2B, GluA1, GluA2, mGluR1 and mGluR5) proteins after 7 days EtOH incubation or after EtOH withdrawal. We found that only GluA1 and mGluR5 expression levels were significantly increased after EtOH withdrawal and, in neuroprotection experiments, we observed that AMPA and mGluR5 antagonists attenuated EtOH withdrawal-induced toxicity. These data suggest that chronic EtOH treatment promotes abnormal synaptic transmission that may lead to CA1 pyramidal cell death after EtOH withdrawal through glutamate receptors and increased excitotoxicity.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy.,Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Elisa Landucci
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Daniele Bani
- Research Unit of Histology and Embryology, Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Flavio Moroni
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Guido Mannaioni
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | | |
Collapse
|
90
|
Prisciandaro JJ, Schacht JP, Prescot AP, Renshaw PF, Brown TR, Anton RF. Brain Glutamate, GABA, and Glutamine Levels and Associations with Recent Drinking in Treatment-Naïve Individuals with Alcohol Use Disorder Versus Light Drinkers. Alcohol Clin Exp Res 2019; 43:221-226. [PMID: 30537347 DOI: 10.1111/acer.13931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/27/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Proton magnetic resonance spectroscopy (1 H-MRS) studies have demonstrated abnormal levels of a variety of neurometabolites in inpatients/outpatients with alcohol use disorder (AUD) following acute alcohol withdrawal relative to healthy controls. In contrast, few studies have compared neurometabolite levels between less severe, treatment-naïve AUD individuals and light drinkers (LD) or related them to recent alcohol consumption. The present study compared neurometabolite levels between treatment-naïve AUD and LD individuals. METHODS Twenty treatment-naïve individuals with AUD and 20 demographically matched LD completed an 1 H-MRS scan, approximately 2.5 days following their last reported drink. 1 H-MRS data were acquired in dorsal anterior cingulate (dACC) using a 2-dimensional J-resolved point-resolved spectroscopy sequence. dACC neurometabolite levels, with a focus on glutamate, glutamine, and GABA, were compared between AUD and LD participants. The associations between metabolite levels and recent drinking were explored. RESULTS AUD participants had significantly lower concentrations of GABA (Cohen's d = 0.79, p = 0.017) and glutamine (Cohen's d = 1.12, p = 0.005), but not glutamate (Cohen's d = 0.05, p = 0.893), relative to LD. As previously reported, AUD participants' glutamate and N-acetylaspartate concentrations were inversely associated with their number of heavy drinking days. In contrast, neither number of drinking (mean p = 0.56) nor heavy drinking (mean p = 0.47) days were associated with metabolite concentrations in LD. CONCLUSIONS The present study demonstrated significantly lower levels of prefrontal γ-aminobutyric acid and glutamine in treatment-naïve individuals with AUD relative to LD. Whether these findings reflect the neurotoxic consequence and/or neuroadaptive response of alcohol consumption versus a predrinking trait, and therefore a more durable neurochemical disturbance, awaits elucidation from longitudinal studies.
Collapse
Affiliation(s)
- James J Prisciandaro
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Joseph P Schacht
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Andrew P Prescot
- Department of Radiology, University of Utah, Salt Lake City, Utah
| | - Perry F Renshaw
- Department of Psychiatry, University of Utah, Salt Lake City, Utah
| | - Truman R Brown
- Department of Radiology, Medical University of South Carolina, Charleston, South Carolina
| | - Raymond F Anton
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
91
|
Qian Z, Wu X, Qiao Y, Shi M, Liu Z, Ren W, Han J, Zheng Q. Downregulation of mGluR2/3 receptors during morphine withdrawal in rats impairs mGluR2/3- and NMDA receptor-dependent long-term depression in the nucleus accumbens. Neurosci Lett 2018; 690:76-82. [PMID: 30315852 DOI: 10.1016/j.neulet.2018.10.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 09/29/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Abstract
Drugs of abuse modify synaptic long-term potentiation and long-term depression (LTD) in the nucleus accumbens, and the impairment of synaptic plasticity in this brain region may be a universal feature of drug addiction. It is unknown whether metabotropic glutamate receptors (mGluRs) play a role in synaptic plasticity induced by drugs such as morphine. The neurochemical, electrophysiological, and Western blotting experiments reported here reveal a novel form of LTD in synapses of the shell region of the nucleus accumbens induced in vivo by low-frequency stimulation of the medial prefrontal cortex. This plasticity required the activation of N-methyl-d-aspartate receptors and mGluR2/3 but not mGluR5. The expression of mGluR2/3 was downregulated during withdrawal from repeated morphine exposure (10 days after the last injection), resulting in impaired low-frequency stimulation-induced LTD. These results indicate that withdrawal-induced mGluR2/3 downregulation alters neural plasticity after morphine exposure, which may be a mechanism contributing to drug addiction.
Collapse
Affiliation(s)
- Zhaoqiang Qian
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China; College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Xiaojie Wu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Yanning Qiao
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Meimei Shi
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Zhiqiang Liu
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Wei Ren
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China
| | - Qiaohua Zheng
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an, Shaanxi, 710062, PR China.
| |
Collapse
|
92
|
Paik SH, Choi MR, Kwak SM, Bang SH, Kim DJ. Decreased Serum Glutamate Levels in Male Adults with Internet Gaming Disorder: A Pilot Study. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2018; 16:276-281. [PMID: 30121977 PMCID: PMC6124868 DOI: 10.9758/cpn.2018.16.3.276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/29/2017] [Accepted: 06/27/2017] [Indexed: 02/03/2023]
Abstract
Objective Alteration in glutamatergic neurotransmission and dopaminergic dysfunction has been implicated in both the initiation and expression of addiction related behaviors. This pilot study was aimed to investigate the serum levels of glutamate and dopamine in adults with internet gaming disorder (IGD). Methods We measured serum levels of glutamate and dopamine in male participants with IGD (n=26) and age-matched healthy controls (n=25). Clinical interviews were performed to identify IGD and to rule out psychiatric comorbidities. Serum levels of glutamate and dopamine were examined by enzyme immunoassays using ELISA Kits. Results Serum levels of glutamate were lower among IGD than control (IGD: 24.184±12.303 μg/ml; control: 33.676±12.413μg/ml; t=2.742, p=0.008), while levels of dopamine did not differ between. Serum glutamate and dopamine levels did not correlate with gaming hours and exposure to game in the IGD group. But serum glutamate levels were positively correlated with the dopamine levels (r=0.360, p=0.013). Conclusion Our results suggest that altered glutamatergic neurotransmission may contribute to the pathophysiology of IGD.
Collapse
Affiliation(s)
- Soo-Hyun Paik
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mi Ran Choi
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Su Min Kwak
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sol Hee Bang
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
93
|
Aboutalebi F, Alaei H, Oryan S. Blockade of Glutamate Receptors within the Prelimbic Cortex Attenuate Concentration of Excitatory Amino Acids in the Morphine Self-administration in Rats. Adv Biomed Res 2018; 7:116. [PMID: 30211129 PMCID: PMC6124215 DOI: 10.4103/abr.abr_121_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background The attitude of research on addiction has been done on the key role of glutamate. As a regard, the prelimbic cortex (PrL) has an important role in addiction, learning, and memory. We tried to investigate the level of glutamate and aspartate concentration after glutamate receptors blockade in this region in the morphine-addicted rats. Materials and Methods In this study, we examined the effects of local infusion of the N-methyl-D-aspartate receptor and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor antagonists, 2-amino-5-phosphonovaleric acid (AP5), and 6-cyano-7-nitroquinoxaline-2, 3-dione (CNQX), into the PrL cortex on the level of excitatory amino acids (EAAs) and glycine. After 11 days of self-administration, the prelimbic area of the brain was taken out, and the EAAs and glycine concentration was measured by high-performance liquid chromatography. Results Morphine resulted in the significant increase in the EAAs concentration within this area (P ≤ 0.001). Microinjection of AP5 into this region before using of morphine significantly decreased the morphine-induced glutamate and aspartate concentration (P ≤ 0.001). CNQX had the same effect and significantly reduced the EAAs concentration compared to the morphine group (P ≤ 0.001). In addition, microinjection of AP5 and CNQX simultaneously increased glycine concentration (P ≤ 0.001). Conclusions These results show that morphine stimulates the EAAs release in the prelimbic area. It seems that microinjection of AP5 or CNQX in this region is effective in reducing morphine-induced EAA. It is suggested that EAA transmission in the PrL cortex may be a possible target for treatment of morphine addiction.
Collapse
Affiliation(s)
- Fateme Aboutalebi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Hojjatallah Alaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
94
|
Wang RY, Chen HJ, Huang CL, Wang JY, Lee TE, Lee HY, Hung CC. Impacts of GRIN3A, GRM6 and TPH2 genetic polymorphisms on quality of life in methadone maintenance therapy population. PLoS One 2018; 13:e0201408. [PMID: 30059533 PMCID: PMC6066242 DOI: 10.1371/journal.pone.0201408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/14/2018] [Indexed: 11/18/2022] Open
Abstract
Opioid addiction is a major public health issue worldwide. Methadone maintenance treatment (MMT) is used to detoxify users of illicit opiates, but drug relapse is common and associated with poor quality of life (QoL). This study investigated the associations between the GRIN3A, GRM6, and TPH2 genetic variants and QoL in the MMT population. A total of 319 participants were included in the study, and genotyping of GRIN3A, GRM6, and TPH2 genes was performed using the Sequenom iPLEX. Associations between genotypes and the domains of QoL were examined through posthoc analysis with LSMEANS syntax using SAS 9.1.3. The single nucleotide polymorphisms rs9325202 and rs1487275 in the TPH2 gene were significantly associated with the QoL domain of physical functioning. The least absolute shrinkage and selection operator regression model revealed that the risk allele rs1487275-G was significantly correlated with the domain of physical functioning when clinical characteristics were considered as covariates. The results of the present study illuminate the importance of the genetic basis of QoL in the MMT population, and suggest that genotypes should be considered as a potential QoL indicator.
Collapse
Affiliation(s)
- Ruey-Yun Wang
- Department of Public Health, China Medical University, Taichung, Taiwan, R.O.C
- Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Hsiu-Ju Chen
- Department of Pharmacy, College of Pharmacy, China Medical University, Taiwan, R.O.C
| | - Chieh-Liang Huang
- Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung, Taiwan, R.O.C
- College of Medicine, China Medical University, Taichung, Taiwan, R.O.C
| | - Jiun-Yi Wang
- Department of Healthcare Administration, Asia University, Wufeng, Taichung, Taiwan, R.O.C
| | - Tsui-Er Lee
- Office of Physical Education, Asia University, Taichung, Taiwan, R.O.C
| | - Hsiang-Yen Lee
- Department of Internal Medicine, Taipei Medical University Hospital, Xinyi District, Taipei City, Taiwan, R.O.C
| | - Chin-Chuan Hung
- Department of Pharmacy, College of Pharmacy, China Medical University, Taiwan, R.O.C
- Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan, R.O.C
- * E-mail:
| |
Collapse
|
95
|
The neurobiology of addiction. A vulnerability/resilience perspective. EUROPEAN JOURNAL OF PSYCHIATRY 2018. [DOI: 10.1016/j.ejpsy.2018.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
96
|
Aboutalebi F, Alaei H, Oryan S, Radahmadi M. Blockade of prelimbic glutamate receptor reduces the reinforcing effect of morphine. Can J Physiol Pharmacol 2018; 96:815-822. [PMID: 29947552 DOI: 10.1139/cjpp-2017-0758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The prelimbic cortex (PrL) as a part of the medial prefrontal cortex (mPFC) plays a crucial role in drug addiction. Previous studies have shown that glutamatergic transmission through the NMDA and AMPA receptors plays an important role in morphine rewarding properties. In this study, we evaluated the effect of glutamate receptors blockade within the PrL on morphine self-administration. Male Wistar rats were randomly selected and divided into 7 groups. Trained rats were placed in self-administration apparatus, where they pressed an active lever for receiving morphine (5 mg/mL) in test groups and saline in saline group during 11 consecutive days for 2 h per session. The effects of intra-prelimbic AMPA receptor antagonist (CNQX; 0.5 and 2.5 μg/0.5 μL) and the NMDA antagonist (AP5; 0.1 and 1 μg/0.5 μL) on self-administration were tested. Our results demonstrated that intra-prelimbic injection of different doses of CNQX and AP5, and co-administration of these 2 drugs before self-administration significantly decreased active lever pressing compared with morphine group (p < 0.001). Also, the number of self-infusion significantly decreased in test groups compared with morphine group (p < 0.001). These findings suggest that a reduction in PrL glutamatergic output can modulate morphine reinforcement.
Collapse
Affiliation(s)
| | - Hojjatallah Alaei
- b Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrbanoo Oryan
- a Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Maryam Radahmadi
- b Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
97
|
Simola N, Brudzynski SM. Rat 50-kHz ultrasonic vocalizations as a tool in studying neurochemical mechanisms that regulate positive emotional states. J Neurosci Methods 2018; 310:33-44. [PMID: 29959002 DOI: 10.1016/j.jneumeth.2018.06.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/22/2018] [Accepted: 06/25/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Adolescent and adult rats emit 50-kHz ultrasonic vocalizations (USVs) to communicate the appetitive arousal and the presence of positive emotional states to conspecifics. NEW METHOD Based on its communicative function, emission of 50-kHz USVs is increasingly being evaluated in preclinical studies of affective behavior, motivation and social behavior. RESULTS Emission of 50-kHz USVs is initiated by the activation of dopamine receptors in the shell subregion of the nucleus accumbens. However, several lines of evidence show that non-dopaminergic receptors may influence the numbers of 50-kHz USVs that are emitted, as well as the acoustic parameters of calls. COMPARISON WITH EXISTING METHODS Emission of 50-kHz USVs is a non-invasive method that may be used to study reward and motivation without the need for extensive training and complex animal manipulations. Moreover, emission of 50-kHz USVs can be used alone or combined with other well-standardized behavioral paradigms (e.g., conditioned place preference, self-administration). CONCLUSIONS This review summarizes the current evidence concerning molecular mechanisms that regulate the emission of 50-kHz USVs. Moreover, the review discusses the usefulness of 50-kHz USVs as an experimental tool to investigate how different neurotransmitter systems regulate the manifestations of positive emotional states, and also use of this tool in preclinical modeling of psychiatric diseases.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Biomedical Sciences, Neuropsychopharmacology Division, University of Cagliari, Italy; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy.
| | - Stefan M Brudzynski
- Department of Psychology, Brock University, St. Catharines, ON, L3 3A1 Canada
| |
Collapse
|
98
|
Barnes SA, Sheffler DJ, Semenova S, Cosford NDP, Bespalov A. Metabotropic Glutamate Receptor 5 as a Target for the Treatment of Depression and Smoking: Robust Preclinical Data but Inconclusive Clinical Efficacy. Biol Psychiatry 2018; 83:955-962. [PMID: 29628194 PMCID: PMC5953810 DOI: 10.1016/j.biopsych.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
The ability of novel pharmacological compounds to improve outcomes in preclinical models is often not translated into clinical efficacy. Psychiatric disorders do not have biological boundaries, and identifying mechanisms to improve the translational bottleneck between preclinical and clinical research domains is an important and challenging task. Glutamate transmission is disrupted in several neuropsychiatric disorders. Metabotropic glutamate (mGlu) receptors represent a diverse class of receptors that contribute to excitatory neurotransmission. Given the wide, yet region-specific manner of expression, developing pharmacological compounds to modulate mGlu receptor activity provides an opportunity to subtly and selectively modulate excitatory neurotransmission. This review focuses on the potential involvement of mGlu5 receptor disruption in major depressive disorder and substance and/or alcohol use disorders. We provide an overview of the justification of targeting mGlu5 receptors in the treatment of these disorders, summarize the preclinical evidence for negatively modulating mGlu5 receptors as a therapeutic target for major depressive disorders and nicotine dependence, and highlight the outcomes of recent clinical trials. While the evidence of mGlu5 receptor negative allosteric modulation has been promising in preclinical investigations, these beneficial effects have not translated into clinical efficacy. In this review, we identify key challenges that may contribute to poor clinical translation and provide suggested approaches moving forward to potentially improve the translation from preclinical to clinical domains. Such approaches may increase the success of clinical trials and may reduce the translational bottleneck that exists in drug discovery for psychiatric disorders.
Collapse
Affiliation(s)
- Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA
| | - Douglas J. Sheffler
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Svetlana Semenova
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive MC 0603, La Jolla, CA 92093, USA,PAREXEL International, 1560 E Chevy Chase Dr, suite 140, Glendale, CA 91206, USA
| | - Nicholas D. P. Cosford
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Anton Bespalov
- EXCIVA, Heidelberg, Germany; Valdman Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia.
| |
Collapse
|
99
|
Leurquin-Sterk G, Ceccarini J, Crunelle CL, Weerasekera A, de Laat B, Himmelreich U, Bormans G, Van Laere K. Cerebral dopaminergic and glutamatergic transmission relate to different subjective responses of acute alcohol intake: an in vivo multimodal imaging study. Addict Biol 2018; 23:931-944. [PMID: 28884874 DOI: 10.1111/adb.12542] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/14/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022]
Abstract
Converging preclinical evidence links extrastriatal dopamine release and glutamatergic transmission via the metabotropic glutamate receptor 5 (mGluR5) to the rewarding properties of alcohol. To date, human evidence is lacking on how and where in the brain these processes occur. Mesocorticolimbic dopamine release upon intravenous alcohol administration and mGluR5 availability were measured in 11 moderate social drinkers by single-session [18 F]fallypride and [18 F]FPEB positron emission tomography, respectively. Additionally, baseline and postalcohol glutamate and glutamine levels in the anterior cingulate cortex (ACC) were measured by using proton-magnetic resonance spectroscopy. To investigate differences in reward domains linked to both neurotransmitters, regional imaging data were related to subjective alcohol responses. Alcohol induced significant [18 F]fallypride displacement in the prefrontal cortex (PFC), temporal and parietal cortices and thalamus (P < 0.05, corrected for multiple comparisons). Dopamine release in the ACC and orbitofrontal and ventromedial PFCs were correlated with subjective 'liking' and 'wanting' effects (P < 0.05). In contrast, baseline mGluR5 availability was positively correlated with the 'high' effect of alcohol in dorsolateral, ventrolateral and ventromedial PFCs and in the medial temporal lobe, thalamus and caudate nucleus (P < 0.05). Although neither proton-magnetic resonance spectroscopy glutamate nor glutamine levels were affected by alcohol, baseline ACC glutamate levels were negatively associated with the alcohol 'liking' effect (P < 0.003). These data reveal new mechanistic understanding and differential neurobiological underpinnings of the effects of acute alcohol consumption on human behavior. Specifically, prefrontal dopamine release may encode alcohol 'liking' and 'wanting' effects in specific areas underlying value processing and motivation, whereas mGluR5 availability in distinct prefrontal-temporal-subcortical regions is more related to the alcohol 'high' effect.
Collapse
Affiliation(s)
- Gil Leurquin-Sterk
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
| | - Jenny Ceccarini
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
| | - Cleo Lina Crunelle
- Toxicological Center, University of Antwerp, Department of Psychiatry; University Hospital Brussels; Belgium
| | | | - Bart de Laat
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
- MoSAIC, Molecular Small Animal Imaging Center; Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology; Biomedical MRI/MoSAIC; Belgium
| | | | - Koen Van Laere
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
- MoSAIC, Molecular Small Animal Imaging Center; Belgium
| |
Collapse
|
100
|
Lebourgeois S, Vilpoux C, Jeanblanc J, Acher F, Marie N, Noble F, Naassila M. Pharmacological activation of mGlu4 and mGlu7 receptors, by LSP2-9166, reduces ethanol consumption and relapse in rat. Neuropharmacology 2018; 133:163-170. [DOI: 10.1016/j.neuropharm.2018.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/12/2018] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
|