51
|
Kruyer A, Kalivas PW. Astrocytes as cellular mediators of cue reactivity in addiction. Curr Opin Pharmacol 2021; 56:1-6. [PMID: 32862045 PMCID: PMC7910316 DOI: 10.1016/j.coph.2020.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023]
Abstract
Relapse to addictive drug use remains a major medical problem worldwide. In rodents, glutamate release in the nucleus accumbens core triggers reinstated drug seeking in response to stress, and drug-associated cues and contexts. Glutamatergic dysregulation in addiction results in part from long-lasting adaptations in accumbens astroglia, including downregulation of the glutamate transporter GLT-1 and retraction from synapses after withdrawal from psychostimulants and opioids. While their capacity to clear glutamate is disrupted by drug use and withdrawal, accumbens astrocytes undergo rapid, transient plasticity in response to drug-associated cues that reinstate seeking. Cued reinstatement of heroin seeking, for example, restores synaptic proximity of astrocyte processes through ezrin phosphorylation, and enhances GLT-1 surface expression. These adaptations limit drug seeking behavior and largely occur on non-overlapping populations of astroglia. Here we review the growing literature supporting a critical role for accumbens astrocytes in modulating glutamate transmission during drug seeking in rodent models of relapse.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, United States.
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, United States
| |
Collapse
|
52
|
Wright WJ, Dong Y. Psychostimulant-Induced Adaptations in Nucleus Accumbens Glutamatergic Transmission. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a039255. [PMID: 31964644 DOI: 10.1101/cshperspect.a039255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Carrying different aspects of emotional and motivational signals, glutamatergic synaptic projections from multiple limbic and paralimbic brain regions converge to the nucleus accumbens (NAc), in which these arousing signals are processed and prioritized for behavioral output. In animal models of drug addiction, some key drug-induced alterations at NAc glutamatergic synapses underlie important cellular and circuit mechanisms that promote subsequent drug taking, seeking, and relapse. With the focus of cocaine, we review changes at NAc glutamatergic synapses that occur after different drug procedures and abstinence durations, and the behavioral impact of these changes.
Collapse
Affiliation(s)
- William J Wright
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| |
Collapse
|
53
|
Stoops WW, Strickland JC, Hays LR, Rayapati AO, Lile JA, Rush CR. Influence of n-acetylcysteine maintenance on the pharmacodynamic effects of oral ethanol. Pharmacol Biochem Behav 2020; 198:173037. [PMID: 32891709 PMCID: PMC7471929 DOI: 10.1016/j.pbb.2020.173037] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE Glutamate systems play an important role in the abuse related effects of alcohol. n-Acetylcysteine, a drug that promotes glutamate homeostasis, attenuates a range of alcohol effects in preclinical models. OBJECTIVES This human laboratory study determined the influence of n-acetylcysteine maintenance on alcohol self-administration using a model predictive of treatment effectiveness, along with the subjective, performance and physiological effects of alcohol. We hypothesized that n-acetylcysteine would attenuate alcohol self-administration, as well as positive subjective effects of alcohol. METHODS Nine subjects with alcohol use disorder completed this within-subjects study. Subjects were maintained on placebo, 1.2 and 2.4 g n-acetylcysteine in random order on an outpatient basis. After five days of maintenance on the target dose, subjects completed overnight inpatient experimental sessions in which the pharmacodynamic effects of alcohol were determined. RESULTS Alcohol produced prototypic effects (e.g., increased breath alcohol concentration, increased ratings of Feel Drink). n-Acetylcysteine did not alter the effects of alcohol. CONCLUSIONS These results indicate that although n-acetylcysteine can safely be combined with alcohol, it does not attenuate the abuse related effects of alcohol and is unlikely to be an effective standalone alcohol use disorder treatment. However, considering study limitations, future work is needed to further understand whether and how n-acetylcysteine might be used as a treatment for alcohol use disorder (e.g., in combination with a behavioral treatment or another pharmacological agent).
Collapse
Affiliation(s)
- William W Stoops
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA; Center on Drug and Alcohol Research, University of Kentucky College of Medicine, 845 Angliana Ave, Lexington, KY 40508, USA.
| | - Justin C Strickland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Abner O Rayapati
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Joshua A Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| | - Craig R Rush
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| |
Collapse
|
54
|
Abstract
Opioid use disorder (OUD) represents a major public health problem that affects millions of people in the USA and worldwide. The relapsing and recurring aspect of OUD, driven by lasting neurobiological adaptations at different reward centres in the brain, represents a major obstacle towards successful long-term remission from opioid use. Currently, three drugs that modulate the function of the opioidergic receptors, methadone, buprenorphine and naltrexone have been approved by the US Food and Drug Administration (FDA) to treat OUD. In this review, we discuss the limitations and challenges associated with the current maintenance and medication-assisted withdrawal strategies commonly used to treat OUD. We further explore the involvement of glutamatergic, endocannabinoid and orexin signaling systems in the development, maintenance and expression of addiction-like behaviours in animal models of opioid addiction, and as potential and novel targets to expand therapeutic options to treat OUD. Despite a growing preclinical literature highlighting the role of these potential targets in animal models of opioid addiction, clinical and translational studies for novel treatments of OUD remain limited and inconclusive. Further preclinical and clinical investigations are needed to expand the arsenal of primary treatment options and adjuncts to maximise efficacy and prevent relapse.
Collapse
|
55
|
Namba MD, Kupchik YM, Spencer SM, Garcia‐Keller C, Goenaga JG, Powell GL, Vicino IA, Hogue IB, Gipson CD. Accumbens neuroimmune signaling and dysregulation of astrocytic glutamate transport underlie conditioned nicotine-seeking behavior. Addict Biol 2020; 25:e12797. [PMID: 31330570 PMCID: PMC7323912 DOI: 10.1111/adb.12797] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
Nicotine self-administration is associated with decreased expression of the glial glutamate transporter (GLT-1) and the cystine-glutamate exchange protein xCT within the nucleus accumbens core (NAcore). N-acetylcysteine (NAC) has been shown to restore these proteins in a rodent model of drug addiction and relapse. However, the specific molecular mechanisms driving its inhibitory effects on cue-induced nicotine reinstatement are unknown. Here, we confirm that extinction of nicotine-seeking behavior is associated with impaired NAcore GLT-1 function and expression and demonstrates that reinstatement of nicotine seeking rapidly enhances membrane fraction GLT-1 expression. Extinction and cue-induced reinstatement of nicotine seeking was also associated with increased tumor necrosis factor alpha (TNFα) and decreased glial fibrillary acidic protein (GFAP) expression in the NAcore. NAC treatment (100 mg/kg/day, i.p., for 5 d) inhibited cue-induced nicotine seeking and suppressed AMPA to NMDA current ratios, suggesting that NAC reduces NAcore postsynaptic excitability. In separate experiments, rats received NAC and an antisense vivo-morpholino to selectively suppress GLT-1 expression in the NAcore during extinction and were subsequently tested for cue-induced reinstatement of nicotine seeking. NAC treatment rescued NAcore GLT-1 expression and attenuated cue-induced nicotine seeking, which was blocked by GLT-1 antisense. NAC also reduced TNFα expression in the NAcore. Viral manipulation of the NF-κB pathway, which is downstream of TNFα, revealed that cue-induced nicotine seeking is regulated by NF-κB pathway signaling in the NAcore independent of GLT-1 expression. Ultimately, these results are the first to show that immunomodulatory mechanisms may regulate known nicotine-induced alterations in glutamatergic plasticity that mediate cue-induced nicotine-seeking behavior.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of PsychologyArizona State University Tempe AZ USA
| | - Yonatan M. Kupchik
- Department of Medical NeurobiologyHebrew University of Jerusalem Jerusalem Israel
| | - Sade M. Spencer
- Department of PharmacologyUniversity of Minnesota Minneapolis MN USA
| | | | | | - Gregory L. Powell
- Department of PsychologyArizona State University Tempe AZ USA
- School of Life SciencesArizona State University Tempe AZ USA
| | - Ian A. Vicino
- School of Life SciencesArizona State University Tempe AZ USA
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign InstituteArizona State University Tempe AZ USA
| | - Ian B. Hogue
- School of Life SciencesArizona State University Tempe AZ USA
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign InstituteArizona State University Tempe AZ USA
| | | |
Collapse
|
56
|
Caffino L, Verheij MMM, Roversi K, Targa G, Mottarlini F, Popik P, Nikiforuk A, Golebiowska J, Fumagalli F, Homberg JR. Hypersensitivity to amphetamine's psychomotor and reinforcing effects in serotonin transporter knockout rats: Glutamate in the nucleus accumbens. Br J Pharmacol 2020; 177:4532-4547. [PMID: 32721055 PMCID: PMC7484509 DOI: 10.1111/bph.15211] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose Amphetamine (AMPH) use disorder is a serious health concern, but, surprisingly, little is known about the vulnerability to the moderate and compulsive use of this psychostimulant and its underlying mechanisms. Previous research showed that inherited serotonin transporter (SERT) down‐regulation increases the motor response to cocaine, as well as moderate (as measured during daily 1‐h self‐administration sessions) and compulsive (as measured during daily 6‐h self‐administration sessions) intake of this psychostimulant. Here, we sought to investigate whether these findings generalize to AMPH and the underlying mechanisms in the nucleus accumbens. Experimental Approach In serotonin transporter knockout (SERT−/−) and wild‐type control (SERT+/+) rats, we assessed the locomotor response to acute AMPH and i.v. AMPH self‐administration under short access (ShA: 1‐h daily sessions) and long access (LgA: 6‐h daily sessions) conditions. Twenty‐four hours after AMPH self‐administration, we analysed the expression of glutamate system components in the nucleus accumbens shell and core. Key Results We found that SERT−/− animals displayed an increased AMPH‐induced locomotor response and increased AMPH self‐administration under LgA but not ShA conditions. Further, we observed changes in the vesicular and glial glutamate transporters, NMDA and AMPA receptor subunits, and their respective postsynaptic scaffolding proteins as function of SERT genotype and AMPH exposure (baseline, ShA, and LgA), specifically in the nucleus accumbens shell. Conclusion and Implications We demonstrate that SERT gene deletion increases the psychomotor and reinforcing effects of AMPH and that the latter is potentially mediated, at least in part, by homeostatic changes in the glutamatergic synapse of the nucleus accumbens shell and/or core.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Michel M M Verheij
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Karine Roversi
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Piotr Popik
- Department of Behavioural Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Agnieska Nikiforuk
- Department of Behavioural Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Joanna Golebiowska
- Department of Behavioural Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
57
|
Ronsley C, Nolan S, Knight R, Hayashi K, Klimas J, Walley A, Wood E, Fairbairn N. Treatment of stimulant use disorder: A systematic review of reviews. PLoS One 2020; 15:e0234809. [PMID: 32555667 PMCID: PMC7302911 DOI: 10.1371/journal.pone.0234809] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS Stimulant use disorder contributes to a substantial worldwide burden of disease, although evidence-based treatment options are limited. This systematic review of reviews aims to: (i) synthesize the available evidence on both psychosocial and pharmacological interventions for the treatment of stimulant use disorder; (ii) identify the most effective therapies to guide clinical practice, and (iii) highlight gaps for future study. METHODS A systematic database search was conducted to identify systematic reviews and meta-analyses. Eligible studies were those that followed standard systematic review methodology and assessed randomized controlled trials focused on the efficacy of interventions for stimulant use disorder. Articles were critically appraised using an assessment tool adapted from Palmeteer et al. and categorized for quality as 'core' or 'supplementary' reviews. Evidence from the included reviews were further synthesized according to pharmacological or non-pharmacological management themes. RESULTS Of 476 identified records, 29 systematic reviews examining eleven intervention modalities were included. The interventions identified include: contingency management, cognitive behavioural therapy, acupuncture, antidepressants, dopamine agonists, antipsychotics, anticonvulsants, disulfiram, opioid agonists, N-Acetylcysteine, and psychostimulants. There was sufficient evidence to support the efficacy of contingency management programs for treatment of stimulant use disorder. Psychostimulants, n-acetylcysteine, opioid agonist therapy, disulfiram and antidepressant pharmacological interventions were found to have insufficient evidence to support or discount their use. Results of this review do not support the use of all other treatment options. CONCLUSIONS The results of this review supports the use of contingency management interventions for the treatment of stimulant use disorder. Although evidence to date is insufficient to support the clinical use of psychostimulants, our results demonstrate potential for future research in this area. Given the urgent need for effective pharmacological treatments for stimulant use disorder, high-quality primary research focused on the role of psychostimulant medications for the treatment of stimulant use disorder is needed.
Collapse
Affiliation(s)
- Claire Ronsley
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Seonaid Nolan
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Rod Knight
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Kanna Hayashi
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Jano Klimas
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Alex Walley
- Department of General Internal Medicine, Boston Medical Center, Boston, MA, United States of America
- Boston University School of Medicine, Boston, MA, United States of America
| | - Evan Wood
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Nadia Fairbairn
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, St. Paul’s Hospital, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
58
|
Hervig ME, Piilgaard L, Božič T, Alsiö J, Robbins TW. Glutamatergic and Serotonergic Modulation of Rat Medial and Lateral Orbitofrontal Cortex in Visual Serial Reversal Learning. ACTA ACUST UNITED AC 2020; 13:438-458. [PMID: 33613854 PMCID: PMC7872199 DOI: 10.1037/pne0000221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
Adapting behavior to a dynamic environment requires both steadiness when the environment is stable and behavioral flexibility in response to changes. Much evidence suggests that cognitive flexibility, which can be operationalized in reversal learning tasks, is mediated by cortico-striatal circuitries, with the orbitofrontal cortex (OFC) playing a prominent role. The OFC is a functionally heterogeneous region, and we have previously reported differential roles of lateral (lOFC) and medial (mOFC) regions in a touchscreen serial visual reversal learning task for rats using pharmacological inactivation. Here, we investigated the effects of pharmacological overactivation of these regions using a glutamate transporter 1 (GLT-1) inhibitor, dihydrokainate (DHK), which increases extracellular glutamate by blocking its reuptake. We also tested the impact of antagonism of the serotonin 2A receptor (5-HT2AR), which modulates glutamate action, in the mOFC and lOFC on the same task. Overactivation induced by DHK produced dissociable effects in the mOFC and lOFC, with more prominent effects in the mOFC, specifically improving performance in the early, perseveration phase. Intra-lOFC DHK increased the number of omitted responses without affecting errors. In contrast, blocking the 5-HT2AR in the lOFC impaired reversal learning overall, while mOFC 5-HT2AR blockade had no effect. These results further support dissociable roles of the rodent mOFC and lOFC in deterministic visual reversal learning and indicate that modulating glutamate transmission through blocking the GLT-1 and the 5-HT2AR have different roles in these two structures. This study further supports dissociable roles of specific orbitofrontal subregions, as well as glutamatergic and serotonergic transmission in these subregions, in cognitive flexibility. This knowledge will add to the understanding of specific neural mechanisms underlying inflexible behaviour across psychiatric disorders.
Collapse
Affiliation(s)
- Mona E Hervig
- Department of Psychology, University of Cambridge, and Department of Neuroscience, University of Copenhagen
| | - Louise Piilgaard
- Department of Psychology, University of Cambridge, and Behavioral and Clinical Neuroscience Institute, University of Cambridge
| | - Tadej Božič
- Department of Psychology, University of Cambridge, and Behavioral and Clinical Neuroscience Institute, University of Cambridge
| | - Johan Alsiö
- Department of Psychology, University of Cambridge, and Behavioral and Clinical Neuroscience Institute, University of Cambridge
| | - Trevor W Robbins
- Department of Psychology, University of Cambridge, and Behavioral and Clinical Neuroscience Institute, University of Cambridge
| |
Collapse
|
59
|
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
|
60
|
Everett NA, Baracz SJ, Cornish JL. The effect of chronic oxytocin treatment during abstinence from methamphetamine self-administration on incubation of craving, reinstatement, and anxiety. Neuropsychopharmacology 2020; 45:597-605. [PMID: 31715618 PMCID: PMC7021828 DOI: 10.1038/s41386-019-0566-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/27/2019] [Accepted: 11/03/2019] [Indexed: 12/22/2022]
Abstract
Methamphetamine (METH) abuse is characterised by chronic relapse and anxiety, for which there are no effective pharmacotherapies. Acute treatment with the neuropeptide oxytocin has shown therapeutic potential for METH addiction and has social and anxiolytic effects in METH-naïve rats. However, the effects of chronic oxytocin treatment in METH-experienced rats are unknown. This study investigated the effects of repeated oxytocin treatment during abstinence from METH self-administration on incubation of cue-induced relapse, yohimbine- and METH-induced reinstatement, trait anxiety, and social interaction. Male and female Sprague-Dawley rats self-administered intravenous METH for 2 h/day (12 days) and then on short-access (2 h/day; ShA) or long-access (6 h/day; LgA) sessions (10 days). Rats underwent 30 days of drug abstinence, during which they received 15 days of intraperitoneal oxytocin (1 mg/kg) or saline (days 6-20) injections. Anxiety and social interaction were tested on days 25-28, and incubation was assessed by testing cue-induced relapse on days 2 and 30. Rats underwent extinction after the final cue-relapse test, followed by yohimbine- and METH-primed reinstatement. LgA, but not ShA rats exhibited incubation of METH-craving and enhanced METH-primed reinstatement in both sexes, and enhanced yohimbine-induced reinstatement in females. Importantly, chronic oxytocin attenuated incubation and METH-primed reinstatement in both sexes, and yohimbine-induced reinstatement in females, although only in LgA rats. LgA produced a heightened anxiety phenotype, which was partially rescued by chronic oxytocin treatment. Using a translatable addiction model, these findings demonstrate the therapeutic efficacy of chronic oxytocin after METH self-administration and supports the clinical utility of oxytocin for METH addiction in both sexes.
Collapse
Affiliation(s)
- Nicholas A. Everett
- 0000 0001 2158 5405grid.1004.5Centre for Emotional Health, Department of Psychology, Macquarie University, Balaclava Road, North Ryde, NSW 2109 Australia
| | - Sarah J. Baracz
- 0000 0001 2158 5405grid.1004.5Centre for Emotional Health, Department of Psychology, Macquarie University, Balaclava Road, North Ryde, NSW 2109 Australia
| | - Jennifer L. Cornish
- 0000 0001 2158 5405grid.1004.5Centre for Emotional Health, Department of Psychology, Macquarie University, Balaclava Road, North Ryde, NSW 2109 Australia
| |
Collapse
|
61
|
Depressive symptoms and cannabis use in a placebo-controlled trial of N-Acetylcysteine for adult cannabis use disorder. Psychopharmacology (Berl) 2020; 237:479-490. [PMID: 31712969 PMCID: PMC7024037 DOI: 10.1007/s00213-019-05384-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
RATIONALE Depression is common among individuals with cannabis use disorder (CUD), particularly individuals who present to CUD treatment. Treatments that consider this comorbidity are essential. OBJECTIVES The goal of this secondary analysis was to examine whether N-acetylcysteine (NAC) reduced depressive symptoms among adults (age 18-50) with CUD (N = 302) and whether the effect of NAC on cannabis cessation varied as a result of baseline levels of depression. Bidirectional associations between cannabis use amount and depression were also examined. METHODS Data for this secondary analysis were from a National Drug Abuse Treatment Clinical Trials Network (NIDA CTN) multi-site clinical trial for CUD. Adults with CUD (N = 302) were randomized to receive 2400 mg of NAC daily or matched placebo for 12 weeks. All participants received abstinence-based contingency management. Cannabis quantity was measured by self-report, and weekly urinary cannabinoid levels (11-nor-9-carboxy-Δ9-tetrahydrocannabinol) confirmed abstinence. Depressive symptoms were measured by the Hospital Anxiety and Depression Scale. RESULTS Depressive symptoms did not differ between the NAC and placebo groups during treatment. There was no significant interaction between treatment and baseline depression predicting cannabis abstinence during treatment. Higher baseline depression was associated with decreased abstinence throughout treatment and a significant gender interaction suggested that this may be particularly true for females. Cross-lagged panel models suggested that depressive symptoms preceded increased cannabis use amounts (in grams) during the subsequent month. The reverse pathway was not significant (i.e., greater cannabis use preceding depressive symptoms). CONCLUSIONS Results from this study suggest that depression may be a risk factor for poor CUD treatment outcome and therefore should be addressed in the context of treatment. However, results do not support the use of NAC to concurrently treat co-occurring depressive symptoms and CUD in adults. TRIAL REGISTRATION Clinicaltrials.gov: NCT01675661.
Collapse
|
62
|
Goenaga J, Powell GL, Leyrer-Jackson JM, Piña J, Phan S, Prakapenka AV, Koebele SV, Namba MD, McClure EA, Bimonte-Nelson HA, Gipson CD. N-acetylcysteine yields sex-specific efficacy for cue-induced reinstatement of nicotine seeking. Addict Biol 2020; 25:e12711. [PMID: 30734439 PMCID: PMC6685767 DOI: 10.1111/adb.12711] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022]
Abstract
Women report greater craving during certain phases of the menstrual cycle. As well, research indicates that pharmacotherapies for smoking may be less efficacious in women compared with men, which may be due to interactions with natural fluctuations in ovarian hormone levels. N-Acetylcysteine (NAC) is a glutamatergic compound that has shown some efficacy in treating substance use disorders and aids in the prevention of relapse. However, it is unclear whether NAC has sex-specific effectiveness for nicotine relapse treatment. Given that NAC has shown promise to reduce nicotine reinstatement in preclinical models using male rats, the exploration of potential sex differences in the efficacy of NAC is warranted. Using a rat model, we first investigated the ability of NAC treatment (100 mg/kg, ip) during nicotine withdrawal with extinction training to reduce cue-induced nicotine seeking in male and female rats. Next, we assessed whether NAC's effects were estrous cycle-dependent for female rats. Results show that following NAC treatment during extinction, reinstatement of nicotine seeking was significantly decreased in males but not females, indicating a sex-specific effect of NAC. Furthermore, for females, both vehicle- and NAC-treated groups significantly reinstated nicotine-seeking behavior compared with extinction, regardless of estrous cycle phase. These results suggest that NAC is inefficacious in reducing nicotine relapse in females regardless of estrous cycle phase at the dose evaluated here. These collective findings could have important clinical implications for use and efficacy of NAC as a pharmacotherapy for freely cycling women smokers.
Collapse
Affiliation(s)
| | | | | | - Jose Piña
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Sandy Phan
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Alesia V. Prakapenka
- Department of Psychology, Arizona State University, Tempe, AZ
- Arizona Alzheimer’s Consortium, Phoenix, AZ
| | - Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, AZ
- Arizona Alzheimer’s Consortium, Phoenix, AZ
| | - Mark D. Namba
- Department of Psychology, Arizona State University, Tempe, AZ
| | - Erin A. McClure
- Department of Psychiatry, Medical University of South Carolina, Charleston, SC
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ
- Arizona Alzheimer’s Consortium, Phoenix, AZ
| | | |
Collapse
|
63
|
Kruyer A, Chioma VC, Kalivas PW. The Opioid-Addicted Tetrapartite Synapse. Biol Psychiatry 2020; 87:34-43. [PMID: 31378302 PMCID: PMC6898767 DOI: 10.1016/j.biopsych.2019.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/17/2022]
Abstract
Opioid administration in preclinical models induces long-lasting adaptations in reward and habit circuitry. The latest research demonstrates that in the nucleus accumbens, opioid-induced excitatory synaptic plasticity involves presynaptic and postsynaptic elements as well as adjacent astroglial processes and the perisynaptic extracellular matrix. We outline opioid-induced modifications within each component of the tetrapartite synapse and provide a neurobiological perspective on how these adaptations converge to produce addiction-related behaviors in rodent models. By incorporating changes observed at each of the excitatory synaptic compartments into a unified framework of opioid-induced glutamate dysregulation, we highlight new avenues for restoring synaptic homeostasis that might limit opioid craving and relapse vulnerability.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Vivian C Chioma
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
64
|
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
|
65
|
Squeglia LM, Fadus MC, McClure EA, Tomko RL, Gray KM. Pharmacological Treatment of Youth Substance Use Disorders. J Child Adolesc Psychopharmacol 2019; 29:559-572. [PMID: 31009234 PMCID: PMC6727439 DOI: 10.1089/cap.2019.0009] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
While the majority of youth who experiment with alcohol and drugs do not develop problematic levels of use, 5% of adolescents and 15% of young adults meet criteria for a substance use disorder (SUD). Pharmacotherapy, in combination with behavioral interventions, has the potential to increase the likelihood of successful treatment for youth struggling with SUD; however, the literature in this area is limited. To date, there are no Food and Drug Administration (FDA)-approved medications for adolescent SUD, other than buprenorphine, which has been approved down to 16 years of age for opioid use disorder. Despite alcohol and cannabis being the most commonly used substances during adolescence, only three medications have been tested among this demographic, and only two have warranted further study (i.e., naltrexone for alcohol and N-acetylcysteine for cannabis use disorder). Although less common in adolescents and young adults, the most promising pharmacological findings for this age group are for opioid (buprenorphine) and tobacco (bupropion and varenicline) use disorders. In addition, despite the recent marked increases in electronic nicotine delivery systems (i.e., vaping) among youth, treatment strategies are still in their infancy and no recommendation exists for how to promote cessation for youth vaping. Current findings are limited by: small, demographically homogeneous samples; few trials, including a substantial number of youth younger than 18; low retention; medication adherence rates; and minimal information on effective dosing levels and long-term outcomes. Overall, pharmacotherapy may be a potentially effective strategy to increase treatment effects; however, more rigorous research trials are warranted before FDA approval would be granted for any of the potential adjunctive medications in this age group.
Collapse
Affiliation(s)
- Lindsay M. Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina.,Address correspondence to: Lindsay M. Squeglia, PhD, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC 29425
| | - Matthew C. Fadus
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Erin A. McClure
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel L. Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin M. Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
66
|
Caffino L, Giannotti G, Messa G, Mottarlini F, Fumagalli F. Repeated cocaine exposure dysregulates BDNF expression and signaling in the mesocorticolimbic pathway of the adolescent rat. World J Biol Psychiatry 2019; 20:531-544. [PMID: 29380665 DOI: 10.1080/15622975.2018.1433328] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objectives: Long-term abstinence following cocaine exposure up-regulates brain-derived neurotrophic factor (BDNF) expression in the mesocorticolimbic pathway. Given the increased vulnerability to drug abuse typical of adolescence, we hypothesized that changes in BDNF expression may become manifest early after the end of cocaine treatment in the adolescent brain.Methods: Rats received cocaine injections from postnatal day 28 (PND28) to PND42 and the mesocorticolimbic expression of BDNF was measured by real-time PCR and Western blotting at PND43.Results: In the ventral tegmental area, BDNF-tropomyosin receptor kinase B (TrΚB) expression and phosphorylation are enhanced while the intracellular signaling is unaltered. In the nucleus accumbens (NAc) shell and core, BDNF and its signaling were down-regulated. In the prelimbic (PL) cortex, we found reduced BDNF expression and increased phosphoprylation of TrΚB, ERK and AKT. In the infralimbic (IL) cortex, increased BDNF expression was coupled with reduced activity and expression of its downstream targets. To evaluate the role of glutamate on BDNF-independent changes, we investigated the expression of the transporter GLT-1 and the activation of the NMDA receptor subunit GluN2B, which were both increased in the PL cortex while reduced in the IL cortex.Conclusions: Our results show that adolescent cocaine exposure modulates BDNF system early after treatment in the mesocorticolimbic pathway, identifying a complex but specific set of changes that could provide clues for treatment.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Giannotti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giulia Messa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
67
|
Lefevre E, Gooch H, Josh P, Alexander S, Eyles DW, Burne TH. Functional and molecular changes in the nucleus accumbens of MK-801-sensitized rats. Behav Pharmacol 2019; 30:383-395. [DOI: 10.1097/fbp.0000000000000447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
68
|
Liu Z, Xu Y, Zhang X, Miao J, Han J, Zhu Z. Riluzole blocks HU210-facilitated ventral tegmental long-term depression by enhancing glutamate uptake in astrocytes. Neurosci Lett 2019; 704:201-207. [DOI: 10.1016/j.neulet.2019.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/09/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
|
69
|
Pinky PD, Bloemer J, Smith WD, Moore T, Hong H, Suppiramaniam V, Reed MN. Prenatal cannabinoid exposure and altered neurotransmission. Neuropharmacology 2019; 149:181-194. [PMID: 30771373 DOI: 10.1016/j.neuropharm.2019.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/18/2019] [Accepted: 02/12/2019] [Indexed: 11/26/2022]
Abstract
Marijuana is one of the most commonly used illicit drugs worldwide. In addition, use of synthetic cannabinoids is increasing, especially among adolescents and young adults. Although human studies have shown that the use of marijuana during pregnancy leads to adverse behavioral effects, such as deficiencies in attention and executive function in affected offspring, the rate of marijuana use among pregnant women is steadily increasing. Various aspects of human behavior including emotion, learning, and memory are dependent on complex interactions between multiple neurotransmitter systems that are especially vulnerable to alterations during the developmental period. Thus, exploration of neurotransmitter changes in response to prenatal cannabinoid exposure is crucial to develop an understanding of how homeostatic imbalance and various long-term neurobehavioral deficits manifest following the abuse of marijuana or other synthetic cannabinoids during pregnancy. Current literature confirms that vast alterations to neurotransmitter systems are present following prenatal cannabinoid exposure, and many of these alterations within the brain are region specific, time-dependent, and sexually dimorphic. In this review, we aim to provide a summary of observed changes to various neurotransmitter systems following cannabinoid exposure during pregnancy and to draw possible correlations to reported behavioral alterations in affected offspring.
Collapse
Affiliation(s)
- Priyanka D Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Warren D Smith
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA
| | - Timothy Moore
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| | - Miranda N Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, USA; Center for Neuroscience Initiative, Auburn University, Auburn, AL, USA.
| |
Collapse
|
70
|
Lebourgeois S, González-Marín MC, Antol J, Naassila M, Vilpoux C. Evaluation of N-acetylcysteine on ethanol self-administration in ethanol-dependent rats. Neuropharmacology 2019; 150:112-120. [DOI: 10.1016/j.neuropharm.2019.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 02/15/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
|
71
|
Yimer EM, Hishe HZ, Tuem KB. Repurposing of the β-Lactam Antibiotic, Ceftriaxone for Neurological Disorders: A Review. Front Neurosci 2019; 13:236. [PMID: 30971875 PMCID: PMC6444273 DOI: 10.3389/fnins.2019.00236] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
To date, there is no cure or disease-modifying agents available for most well-known neurological disorders. Current therapy is typically focused on relieving symptoms and supportive care in improving the quality of life of affected patients. Furthermore, the traditional de novo drug discovery technique is more challenging, particularly for neurological disorders. Therefore, the repurposing of existing drugs for these conditions is believed to be an efficient and dynamic approach that can substantially reduce the investments spent on drug development. Currently, there is emerging evidence that suggests the potential effect of a beta-lactam antibiotic, ceftriaxone (CEF), to alleviate the symptoms of different experimentally-induced neurological disorders: Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, epileptic-seizure, brain ischemia, traumatic brain injuries, and neuropathic pain. CEF also affects the markers of oxidative status and neuroinflammation, glutamatergic systems as well as various aggregated toxic proteins involved in the pathogenesis of different neurological disorders. Moreover, it was found that CEF administration to drug dependent animal models improved the withdrawal symptoms upon drug discontinuation. Thus, this review aimed to describe the effects of CEF against multiple models of neurological illnesses, drug dependency, and withdrawal. It also emphasizes the possible mechanisms of neuroprotective actions of CEF with respective neurological maladies.
Collapse
Affiliation(s)
- Ebrahim M Yimer
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Hailemichael Zeru Hishe
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Kald Beshir Tuem
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
72
|
Siemsen BM, Reichel CM, Leong KC, Garcia-Keller C, Gipson CD, Spencer S, McFaddin JA, Hooker KN, Kalivas PW, Scofield MD. Effects of Methamphetamine Self-Administration and Extinction on Astrocyte Structure and Function in the Nucleus Accumbens Core. Neuroscience 2019; 406:528-541. [PMID: 30926546 PMCID: PMC6545487 DOI: 10.1016/j.neuroscience.2019.03.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 01/01/2023]
Abstract
Astrocytes provide support for neurons, regulate metabolic processes, and influence neuronal communication in a variety of ways, including through the homeostatic regulation of glutamate. Following 2-h cocaine or methamphetamine self-administration (SA) and extinction, rodents display decreased levels of basal glutamate in the nucleus accumbens core (NAcore), which transitions to elevated glutamate levels during drug seeking. We hypothesized that, like cocaine, this glutamate 'overflow' during methamphetamine seeking arises via decreased expression of the astroglial glutamate transporter GLT-1, and withdrawal of perisynaptic astroglial processes (PAPs) from synapses. As expected, methamphetamine self-administration and extinction decreased the level of contact made by PAPs in the NAcore, yet did not impact glutamate uptake, GLT-1 expression, or the general structural characteristics of astrocytes. Interestingly, systemic administration of N-acetylcysteine (NAC), a drug that both upregulates GLT-1 and promotes glial-glutamate release, reduced cued methamphetamine seeking. In order to test the impact of astrocyte activation and the induction of glial glutamate release within the NAcore, we employed astrocyte-specific expression of designer receptors exclusively activated by designer drugs (DREADDs). We show here that acute activation of Gq-coupled DREADDs in this region inhibited cued methamphetamine seeking. Taken together, these data indicate that cued methamphetamine seeking following two-hour SA is not mediated by deficient glutamate clearance in the NAcore, yet can be inhibited by engaging NAcore astrocytes.
Collapse
Affiliation(s)
- B M Siemsen
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - C M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - K C Leong
- Department of Psychology, Trinity University, San Antonio, TX, USA
| | - C Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - C D Gipson
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - S Spencer
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - J A McFaddin
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - K N Hooker
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - M D Scofield
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
73
|
Ma B, Mei D, Wang F, Liu Y, Zhou W. Cognitive enhancers as a treatment for heroin relapse and addiction. Pharmacol Res 2019; 141:378-383. [DOI: 10.1016/j.phrs.2019.01.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/22/2018] [Accepted: 01/14/2019] [Indexed: 12/31/2022]
|
74
|
Powell GL, Leyrer‐Jackson JM, Goenaga J, Namba MD, Piña J, Spencer S, Stankeviciute N, Schwartz D, Allen NP, Del Franco AP, McClure EA, Olive MF, Gipson CD. Chronic treatment with N-acetylcysteine decreases extinction responding and reduces cue-induced nicotine-seeking. Physiol Rep 2019; 7:e13958. [PMID: 30632301 PMCID: PMC6328917 DOI: 10.14814/phy2.13958] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/25/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022] Open
Abstract
N-acetylcysteine (NAC), a promising glutamatergic therapeutic agent, has shown some clinical efficacy in reducing nicotine use in humans and has been shown to reverse drug-induced changes in glutamatergic neurophysiology. In rats, nicotine-seeking behavior is associated with alterations in glutamatergic plasticity within the nucleus accumbens core (NAcore). Specifically, cue-induced nicotine-seeking is associated with rapid, transient synaptic plasticity (t-SP) in glutamatergic synapses on NAcore medium spiny neurons. The goal of the present study was to determine if NAC reduces nicotine-seeking behavior and reverses reinstatement-associated NAcore glutamatergic alterations. Rats were extinguished from nicotine self-administration, followed by subchronic NAC administration (0 or 100 mg/kg/d) for 4 days prior to cue-induced reinstatement. NAcore synaptic potentiation was measured via dendritic spine morphology and mRNA and protein of relevant glutamatergic genes were quantified. Nicotine-seeking behavior was not reduced by subchronic NAC treatment. Also, NAcore transcript and protein expression of multiple glutamatergic genes, as well as spine morphological measures, were unaffected by subchronic NAC. Finally, chronic NAC treatment (15 days total) during extinction and prior to reinstatement significantly decreased extinction responding and reduced reinstatement of nicotine-seeking compared to vehicle. Together, these results suggest that chronic NAC treatment is necessary for its therapeutic efficacy as a treatment strategy for nicotine addiction and relapse.
Collapse
Affiliation(s)
- Gregory L. Powell
- Department of PsychologyArizona State UniversityTempeArizona
- School of Life SciencesArizona State UniversityTempeArizona
| | | | | | - Mark D. Namba
- Department of PsychologyArizona State UniversityTempeArizona
| | - Jose Piña
- Department of PsychologyArizona State UniversityTempeArizona
| | - Sade Spencer
- Department of NeuroscienceMedical University of South CarolinaCharlestonSC
| | | | - Danielle Schwartz
- Department of NeuroscienceMedical University of South CarolinaCharlestonSC
| | - Nicholas P. Allen
- School of Dental MedicineLake Erie College of Osteopathic MedicineBradentonFlorida
| | | | - Erin A. McClure
- Department of PsychiatryMedical University of South CarolinaCharlestonSouth Carolina
| | | | | |
Collapse
|
75
|
Kim R, Healey KL, Sepulveda-Orengo MT, Reissner KJ. Astroglial correlates of neuropsychiatric disease: From astrocytopathy to astrogliosis. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:126-146. [PMID: 28989099 PMCID: PMC5889368 DOI: 10.1016/j.pnpbp.2017.10.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/24/2017] [Accepted: 10/04/2017] [Indexed: 01/22/2023]
Abstract
Complex roles for astrocytes in health and disease continue to emerge, highlighting this class of cells as integral to function and dysfunction of the nervous system. In particular, escalating evidence strongly implicates a range of changes in astrocyte structure and function associated with neuropsychiatric diseases including major depressive disorder, schizophrenia, and addiction. These changes can range from astrocytopathy, degeneration, and loss of function, to astrogliosis and hypertrophy, and can be either adaptive or maladaptive. Evidence from the literature indicates a myriad of changes observed in astrocytes from both human postmortem studies as well as preclinical animal models, including changes in expression of glial fibrillary protein, as well as changes in astrocyte morphology and astrocyte-mediated regulation of synaptic function. In this review, we seek to provide a comprehensive assessment of these findings and consequently evidence for common themes regarding adaptations in astrocytes associated with neuropsychiatric disease. While results are mixed across conditions and models, general findings indicate decreased astrocyte cellular features and gene expression in depression, chronic stress and anxiety, but increased inflammation in schizophrenia. Changes also vary widely in response to different drugs of abuse, with evidence reflective of features of astrocytopathy to astrogliosis, varying across drug classes, route of administration and length of withdrawal.
Collapse
Affiliation(s)
- Ronald Kim
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kati L Healey
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Marian T Sepulveda-Orengo
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, CB 3270, UNC Chapel Hill, Chapel Hill, NC 27599, United States..
| |
Collapse
|
76
|
Drug addiction: a curable mental disorder? Acta Pharmacol Sin 2018; 39:1823-1829. [PMID: 30382181 DOI: 10.1038/s41401-018-0180-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Drug addiction is a chronic, relapsing brain disorder. Multiple neural networks in the brain including the reward system (e.g., the mesocorticolimbic system), the anti-reward/stress system (e.g., the extended amygdala), and the central immune system, are involved in the development of drug addiction and relapse after withdrawal from drugs of abuse. Preclinical and clinical studies have demonstrated that it is promising to control drug addiction by pharmacologically targeting the addiction-related systems in the brain. Here we review the pharmacological targets within the dopamine system, glutamate system, trace amine system, anti-reward system, and central immune system, which are of clinical interests. Furthermore, we discuss other potential therapies, e.g., brain stimulation, behavioral treatments, and therapeutic gene modulation, which could be effective to treat drug addiction. We conclude that, although drug addiction is a complex disorder that involves complicated neural mechanisms and psychological processes, this mental disorder is treatable and may be curable by therapies such as gene modulation in the future.
Collapse
|
77
|
Tian SW, Yu XD, Cen L, Xiao ZY. Glutamate transporter GLT1 inhibitor dihydrokainic acid impairs novel object recognition memory performance in mice. Physiol Behav 2018; 199:28-32. [PMID: 30389478 DOI: 10.1016/j.physbeh.2018.10.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022]
Abstract
Glutamate transporter GLT1 mediates glutamate uptake, and maintains glutamate homeostasis in the synaptic cleft. Previous studies suggest that blockade of glutamate uptake affects synaptic transmission and plasticity. However, the effect of GLT1 blockade on learning and memory still receives little attention. In the present study, we examined the effect of unilateral intracerebroventricular injection of dihydrokainic acid (DHK), a GLT-1 inhibitor, on novel object recognition (NOR) memory performance. The NOR task involved three sessions including habituation, sampling and test. In experiment 1, DHK injection 0.5 h pre-sampling impaired short-term NOR memory performance. In experiment 2, DHK injection 0.5 h pre-sampling impaired long-term NOR memory acquisition. In experiment 3, DHK injection immediately but not 6 h post-sampling impaired long-term NOR memory consolidation. In experiment 4, DHK injection 0.5 h pre-test impaired long-term NOR memory retrieval. Furthermore, DHK-induced memory performance impairment was not due to its effects on nonspecific responses such as locomotor activity and exploratory behavior. The current findings further extend previous studies on the effects of disruption of glutamate homeostasis on learning and memory.
Collapse
Affiliation(s)
- Shao-Wen Tian
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.
| | - Xu-Dong Yu
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.
| | - Lian Cen
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Zhi-Yong Xiao
- Department of Anesthesiology, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
78
|
Linker KE, Cross SJ, Leslie FM. Glial mechanisms underlying substance use disorders. Eur J Neurosci 2018; 50:2574-2589. [PMID: 30240518 DOI: 10.1111/ejn.14163] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/28/2022]
Abstract
Addiction is a devastating disorder that produces persistent maladaptive changes to the central nervous system, including glial cells. Although there is an extensive body of literature examining the neuronal mechanisms of substance use disorders, effective therapies remain elusive. Glia, particularly microglia and astrocytes, have an emerging and meaningful role in a variety of processes beyond inflammation and immune surveillance, and may represent a promising therapeutic target. Indeed, glia actively modulate neurotransmission, synaptic connectivity and neural circuit function, and are critically poised to contribute to addictive-like brain states and behaviors. In this review, we argue that glia influence the cellular, molecular, and synaptic changes that occur in neurons following drug exposure, and that this cellular relationship is critically modified following drug exposure. We discuss direct actions of abused drugs on glial function through immune receptors, such as Toll-like receptor 4, as well as other mechanisms. We highlight how drugs of abuse affect glia-neural communication, and the profound effects that glial-derived factors have on neuronal excitability, structure, and function. Recent research demonstrates that glia have brain region-specific functions, and glia in different brain regions have distinct contributions to drug-associated behaviors. We will also evaluate the evidence demonstrating that glial activation is essential for drug reward and drug-induced dopamine release, and highlight clinical evidence showing that glial mechanisms contribute to drug abuse liability. In this review, we synthesize the extensive evidence that glia have a unique, pivotal, and underappreciated role in the development and maintenance of addiction.
Collapse
Affiliation(s)
- K E Linker
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - S J Cross
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - F M Leslie
- Department of Pharmacology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
79
|
Neuhofer D, Kalivas P. Metaplasticity at the addicted tetrapartite synapse: A common denominator of drug induced adaptations and potential treatment target for addiction. Neurobiol Learn Mem 2018; 154:97-111. [PMID: 29428364 PMCID: PMC6112115 DOI: 10.1016/j.nlm.2018.02.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/26/2018] [Accepted: 02/07/2018] [Indexed: 11/22/2022]
Abstract
In light of the current worldwide addiction epidemic, the need for successful therapies is more urgent than ever. Although we made substantial progress in our basic understanding of addiction, reliable therapies are lacking. Since 40-60% of patients treated for substance use disorder return to active substance use within a year following treatment discharge, alleviating the vulnerability to relapse is regarded as the most promising avenue for addiction therapy. Preclinical addiction research often focuses on maladaptive synaptic plasticity within the reward pathway. However, drug induced neuroadaptations do not only lead to a strengthening of distinct drug associated cues and drug conditioned behaviors, but also seem to increase plasticity thresholds for environmental stimuli that are not associated with the drug. This form of higher order plasticity, or synaptic metaplasticity, is not expressed as a change in the efficacy of synaptic transmission but as a change in the direction or degree of plasticity induced by a distinct stimulation pattern. Experimental addiction research has demonstrated metaplasticity after exposure to multiple classes of addictive drugs. In this review we will focus on the concept of synaptic metaplasticity in the context of preclinical addiction research. We will take a closer look at the tetrapartite glutamatergic synapse and outline forms of metaplasticity that have been described at the addicted synapse. Finally we will discuss the different potential avenues for pharmacotherapies that target glutamatergic synaptic plasticity and metaplasticity. Here we will argue that aberrant metaplasticity renders the reward seeking circuitry more rigid and hence less able to adapt to changing environmental contingencies. An understanding of the molecular mechanisms that underlie this metaplasticity is crucial for the development of new strategies for addiction therapy. The correction of drug-induced metaplasticity could be used to support behavioral and pharmacotherapies for the treatment of addiction.
Collapse
Affiliation(s)
- Daniela Neuhofer
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States
| |
Collapse
|
80
|
Tomko RL, Gilmore AK, Gray KM. The role of depressive symptoms in treatment of adolescent cannabis use disorder with N-Acetylcysteine. Addict Behav 2018; 85:26-30. [PMID: 29803870 DOI: 10.1016/j.addbeh.2018.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 05/19/2018] [Indexed: 01/09/2023]
Abstract
Relative to adults, adolescents are at greater risk of developing a cannabis use disorder (CUD) and risk may be exacerbated by co-occurring depressive symptoms. N-Acetylcysteine (NAC), an over-the-counter antioxidant, is thought to normalize glutamate transmission. Oxidative stress and glutamate transmission are disrupted in both depression and CUD. Thus, NAC may be particularly effective at promoting cannabis abstinence among adolescents with elevated depressive symptoms. Secondary analyses were conducted using a sub-sample of adolescents with CUD (N = 74) who participated in an 8-week randomized placebo-controlled clinical trial examining the efficacy of NAC for cannabis cessation. It was hypothesized that NAC would reduce severity of depressive symptoms, and that decreases depressive symptom severity would mediate decreases in positive weekly urine cannabinoid tests (11-nor-9-carboxy-Δ9-tetrahydrocannabinol). Additionally, it was expected that adolescents with greater severity of baseline depressive symptoms would be more likely to become abstinent when assigned NAC relative to placebo. Results from linear mixed models and generalized estimating equations did not suggest that NAC reduced severity of depressive symptoms, and the hypothesis that NAC's effect on cannabis cessation would be mediated by reduced depressive symptoms was not supported. However, an interaction between treatment condition and baseline severity of depressive symptoms as a predictor of weekly urine cannabinoid tests was significant, suggesting that NAC was more effective at promoting abstinence among adolescents with heightened baseline depressive symptoms. These secondary findings, though preliminary, suggest a need for further examination of the role of depressive symptoms in treatment of adolescent CUD with NAC.
Collapse
|
81
|
Womersley JS, Townsend DM, Kalivas PW, Uys JD. Targeting redox regulation to treat substance use disorder using N‐acetylcysteine. Eur J Neurosci 2018; 50:2538-2551. [PMID: 30144182 DOI: 10.1111/ejn.14130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/28/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022]
Abstract
Substance use disorder (SUD) is a chronic relapsing disorder characterized by transitioning from acute drug reward to compulsive drug use. Despite the heavy personal and societal burden of SUDs, current treatments are limited and unsatisfactory. For this reason, a deeper understanding of the mechanisms underlying addiction is required. Altered redox status, primarily due to drug-induced increases in dopamine metabolism, is a unifying feature of abused substances. In recent years, knowledge of the effects of oxidative stress in the nervous system has evolved from strictly neurotoxic to include a more nuanced role in redox-sensitive signaling. More specifically, S-glutathionylation, a redox-sensitive post-translational modification, has been suggested to influence the response to drugs of abuse. In this review we will examine the evidence for redox-mediating drugs as therapeutic tools focusing on N-acetylcysteine as a treatment for cocaine addiction. We will conclude by suggesting future research directions that may further advance this field.
Collapse
Affiliation(s)
- Jacqueline S Womersley
- Department of Cellular and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 409 Drug Discovery Building, 70 President Street, Charleston, SC, 29425, USA
| | - Danyelle M Townsend
- Department of Drug Discover and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Joachim D Uys
- Department of Cellular and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 409 Drug Discovery Building, 70 President Street, Charleston, SC, 29425, USA
| |
Collapse
|
82
|
Gibson CL, Codreanu SG, Schrimpe-Rutledge AC, Retzlaff CL, Wright J, Mortlock DP, Sherrod SD, McLean JA, Blakely RD. Global untargeted serum metabolomic analyses nominate metabolic pathways responsive to loss of expression of the orphan metallo β-lactamase, MBLAC1. Mol Omics 2018; 14:142-155. [PMID: 29868674 PMCID: PMC6015503 DOI: 10.1039/c7mo00022g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The C. elegans gene swip-10 encodes an orphan metallo β-lactamase that genetic studies indicate is vital for limiting neuronal excitability and viability. Sequence analysis indicates that the mammalian gene Mblac1 is the likely ortholog of swip-10, with greatest sequence identity localized to the encoded protein's single metallo β-lactamase domain. The substrate for the SWIP-10 protein remains unknown and to date no functional roles have been ascribed to MBLAC1, though we have shown that the protein binds the neuroprotective β-lactam antibiotic, ceftriaxone. To gain insight into the functional role of MBLAC1 in vivo, we used CRISPR/Cas9 methods to disrupt N-terminal coding sequences of the mouse Mblac1 gene, resulting in a complete loss of protein expression in viable, homozygous knockout (KO) animals. Using serum from both WT and KO mice, we performed global, untargeted metabolomic analyses, resolving small molecules via hydrophilic interaction chromatography (HILIC) based ultra-performance liquid chromatography, coupled to mass spectrometry (UPLC-MS/MS). Unsupervised principal component analysis reliably segregated the metabolomes of MBLAC1 KO and WT mice, with 92 features subsequently nominated as significantly different by ANOVA, and for which we made tentative and putative metabolite assignments. Bioinformatic analyses of these molecules nominate validated pathways subserving bile acid biosynthesis and linoleate metabolism, networks known to be responsive to metabolic and oxidative stress. Our findings lead to hypotheses that can guide future targeted studies seeking to identify the substrate for MBLAC1 and how substrate hydrolysis supports the neuroprotective actions of ceftriaxone.
Collapse
Affiliation(s)
- Chelsea L. Gibson
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Simona G. Codreanu
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Alexandra C. Schrimpe-Rutledge
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Cassandra L. Retzlaff
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
| | - Jane Wright
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Doug P. Mortlock
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN USA
| | - Stacy D. Sherrod
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter FL, USA
- Brain Institute, Florida Atlantic University, Jupiter FL, USA
| |
Collapse
|
83
|
Tomko RL, Jones JL, Gilmore AK, Brady KT, Back SE, Gray KM. N-acetylcysteine: A potential treatment for substance use disorders. CURRENT PSYCHIATRY 2018; 17:30-55. [PMID: 30016376 PMCID: PMC5993450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Rachel L Tomko
- Research Assistant Professor, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer L Jones
- Resident Physician, Departments of Psychiatry and Behavioral Sciences and Internal Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Amanda K Gilmore
- Assistant Professor, National Crime Victims Research & Treatment Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Kathleen T Brady
- Distinguished University Professor, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Sudie E Back
- Professor, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin M Gray
- Professor, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
84
|
Riluzole Impairs Cocaine Reinstatement and Restores Adaptations in Intrinsic Excitability and GLT-1 Expression. Neuropsychopharmacology 2018; 43:1212-1223. [PMID: 28990593 PMCID: PMC5916346 DOI: 10.1038/npp.2017.244] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/29/2017] [Accepted: 10/01/2017] [Indexed: 01/01/2023]
Abstract
Adaptations in glutamate signaling within the brain's reward circuitry are observed following withdrawal from several abused drugs, including cocaine. These include changes in intrinsic cellular excitability, glutamate release, and glutamate uptake. Pharmacological or optogenetic reversal of these adaptations have been shown to reduce measures of cocaine craving and seeking, raising the hypothesis that regulation of glutamatergic signaling represents a viable target for the treatment of substance use disorders. Here, we tested the hypothesis that administration of the compound riluzole, which regulates glutamate dynamics in several ways, would reduce cocaine seeking in the rat self-administration and reinstatement model of addiction. Riluzole dose-dependently inhibited cue- and cocaine-primed reinstatement to cocaine, but did not affect locomotor activity or reinstatement to sucrose seeking. Moreover, riluzole reversed bidirectional cocaine-induced adaptations in intrinsic excitability of prelimbic (PL) and infralimbic (IL) pyramidal neurons; a cocaine-induced increase in PL excitability was decreased by riluzole, and a cocaine-induced decrease in IL excitability was increased to normal levels. Riluzole also reversed the cocaine-induced suppression of the high-affinity glutamate transporter 1 (EAAT2/GLT-1) in the nucleus accumbens (NAc). GLT-1 is responsible for the majority of glutamate uptake in the brain, and has been previously reported to be downregulated by cocaine. These results demonstrate that riluzole impairs cocaine reinstatement while rectifying several cellular adaptations in glutamatergic signaling within the brain's reward circuitry, and support the hypothesis that regulators of glutamate homeostasis represent viable candidates for pharmacotherapeutic treatment of psychostimulant relapse.
Collapse
|
85
|
Fischer KD, Houston ACW, Desai RI, Doyle MR, Bergman J, Mian M, Mannix R, Sulzer DL, Choi SJ, Mosharov EV, Hodgson NW, Bechtholt A, Miczek KA, Rosenberg PA. Behavioral phenotyping and dopamine dynamics in mice with conditional deletion of the glutamate transporter GLT-1 in neurons: resistance to the acute locomotor effects of amphetamine. Psychopharmacology (Berl) 2018; 235:1371-1387. [PMID: 29468294 PMCID: PMC5999338 DOI: 10.1007/s00213-018-4848-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022]
Abstract
RATIONALE GLT-1 is the major glutamate transporter in the brain and is expressed predominantly in astrocytes but is also present in excitatory axon terminals. To understand the functional significance of GLT-1 expressed in neurons, we generated a conditional GLT-1 knockout mouse and inactivated GLT-1 in neurons using Cre-recombinase expressed under the synapsin 1 promoter, (synGLT-1 KO). OBJECTIVES Abnormalities of glutamate homeostasis have been shown to affect hippocampal-related behaviors including learning and memory as well as responses to drugs of abuse. Here, we asked whether deletion of GLT-1 specifically from neurons would affect behaviors that assessed locomotor activity, cognitive function, sensorimotor gating, social interaction, as well as amphetamine-stimulated locomotor activity. METHODS/RESULTS We found that the neuronal GLT-1 KO mice performed similarly to littermate controls in the behavioral tests we studied. Although performance in open field testing was normal, the acute locomotor response to amphetamine was significantly blunted in the synGLT-1 KO (40% of control). We found no change in amphetamine-stimulated extracellular dopamine in the medial shell of the nucleus accumbens, no change in electrically stimulated or amphetamine-induced dopamine release, and no change in dopamine tissue content. CONCLUSIONS These results support the view that GLT-1 expression in neurons is required for amphetamine-induced behavioral activation, and suggest that this phenotype is not produced through a change in dopamine uptake or release. Although GLT-1 is highly expressed in neurons in the CA3 region of the hippocampus, the tests used in this study were not able to detect a behavioral phenotype referable to hippocampal dysfunction.
Collapse
Affiliation(s)
- Kathryn D Fischer
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Alex C W Houston
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rajeev I Desai
- Preclinical Pharmacology Program, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Michelle R Doyle
- Preclinical Pharmacology Program, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Jack Bergman
- Preclinical Pharmacology Program, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Maha Mian
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Rebekah Mannix
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - David L Sulzer
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | - Se Joon Choi
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | - Eugene V Mosharov
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | - Nathaniel W Hodgson
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Anita Bechtholt
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Klaus A Miczek
- Departments of Psychiatry, Pharmacology, and Neuroscience, Tufts University, Boston, MA, 02111, USA
| | - Paul A Rosenberg
- Department of Neurology and the F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
86
|
Squeglia LM, Tomko RL, Baker NL, McClure EA, Book GA, Gray KM. The effect of N-acetylcysteine on alcohol use during a cannabis cessation trial. Drug Alcohol Depend 2018; 185:17-22. [PMID: 29413434 PMCID: PMC5889716 DOI: 10.1016/j.drugalcdep.2017.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/27/2017] [Accepted: 12/10/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Individuals with alcohol use disorder (AUD) do not always respond to currently available treatments, and evaluation of new candidate pharmacotherapies is indicated. N-acetylcysteine (NAC), an over-the-counter supplement, has shown promise in treating a variety of substance use disorders, but little research has evaluated its merits as a treatment for AUD. This secondary analysis from the National Drug Abuse Treatment Clinical Trials Network examined the effects of NAC versus placebo on alcohol use among participants with cannabis use disorder (CUD) enrolled in a 12-week, multi-site cannabis cessation trial. METHODS Participants (N = 302, ages 18-50) were randomized to double-blind NAC (1200 mg, twice daily) or placebo. Neither alcohol use nor desire for alcohol cessation were requirements for participation. Participants that returned for at least one treatment visit and had recorded alcohol use data (i.e., total drinks per week, drinking days per week, and binge drinking days per week) were included in the analysis (n = 277). RESULTS Compared to the placebo group, participants in the NAC group had increased odds of between-visit alcohol abstinence [OR = 1.37; 95% CI = 1.06-1.78; p = 0.019], fewer drinks per week [RR = 0.67; 95% CI = 0.48-0.99; p = 0.045], and fewer drinking days per week [RR = 0.69; 95% CI = 0.51-0.92; p = 0.014]. Changes in concurrent cannabis use amounts were not correlated to any of the alcohol use variables. DISCUSSION These findings indicate that NAC may be effective at reducing consumption of alcohol by ∼30% among treatment-seeking adults with CUD, suggesting a need for further trials focused on the effects of NAC on alcohol consumption among individuals seeking treatment for AUD.
Collapse
Affiliation(s)
- Lindsay M Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC, United States.
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC, United States
| | - Nathaniel L Baker
- Department of Public Health Sciences, Medical University of South Carolina, 135 Cannon Street, Suite 303, MSC 835, Charleston, SC, United States
| | - Erin A McClure
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC, United States
| | - George A Book
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC, United States
| | - Kevin M Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President Street, MSC 861, Charleston, SC, United States
| |
Collapse
|
87
|
Ezquer F, Morales P, Quintanilla ME, Santapau D, Lespay-Rebolledo C, Ezquer M, Herrera-Marschitz M, Israel Y. Intravenous administration of anti-inflammatory mesenchymal stem cell spheroids reduces chronic alcohol intake and abolishes binge-drinking. Sci Rep 2018; 8:4325. [PMID: 29567966 PMCID: PMC5864829 DOI: 10.1038/s41598-018-22750-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic alcohol intake leads to neuroinflammation and astrocyte dysfunction, proposed to perpetuate alcohol consumption and to promote conditioned relapse-like binge drinking. In the present study, human mesenchymal stem cells (MSCs) were cultured in 3D-conditions to generate MSC-spheroids, which greatly increased MSCs anti-inflammatory ability and reduced cell volume by 90% versus conventionally 2D-cultured MSCs, enabling their intravenous administration and access to the brain. It is shown, in an animal model of chronic ethanol intake and relapse-drinking, that both the intravenous and intra-cerebroventricular administration of a single dose of MSC-spheroids inhibited chronic ethanol intake and relapse-like drinking by 80–90%, displaying significant effects over 3–5 weeks. The MSC-spheroid administration fully normalized alcohol-induced neuroinflammation, as shown by a reduced astrocyte activation, and markedly increased the levels of the astrocyte Na-glutamate (GLT-1) transporter. This research suggests that the intravenous administration of MSC-spheroids may constitute an effective new approach for the treatment of alcohol-use disorders.
Collapse
Affiliation(s)
- Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
| |
Collapse
|
88
|
Lebourgeois S, González-Marín MC, Jeanblanc J, Naassila M, Vilpoux C. Effect of N-acetylcysteine on motivation, seeking and relapse to ethanol self-administration. Addict Biol 2018; 23:643-652. [PMID: 28557352 DOI: 10.1111/adb.12521] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/10/2017] [Accepted: 04/11/2017] [Indexed: 02/06/2023]
Abstract
Alcohol use disorder is a chronic and highly relapsing disorder, characterized by a loss of control over alcohol consumption and craving. Several studies suggest a key role of glutamate in this disorder. In recent years, the modulation of cystine/glutamate exchange via the xc- system has emerged as a new therapeutic alternative for reducing the excitatory glutamatergic transmission observed after ethanol self-administration in both rats and humans. The objective of this study was to determine whether a treatment with N-acetylcysteine (NAC), a cystine prodrug, could reduce ethanol self-administration, ethanol-seeking behavior and reacquisition of ethanol self-administration. Male Long Evans rats were trained to self-administer 20 percent ethanol in operant cages for several weeks. Once the consumption surpassed 1 g of ethanol/kg body weight/15 minutes, the effect of an acute intraperitoneal injection of NAC (0, 25, 50 or 100 mg/kg) 1 hour before the beginning of each test was evaluated on different aspects of the operant self-administration behavior. We demonstrated antimotivational properties of NAC (100 mg/kg), as ethanol-reinforced responding was reduced in a fixed ratio (-35 percent) and in a progressive ratio schedule (-81 percent). NAC also reduced ethanol-seeking behavior (-77 percent) evaluated as extinction responding in a single extinction session. NAC was able to reduce reacquisition in rats that were abstinent for 17 days, while NAC had no effect on ethanol relapse in rats previously exposed to six extinction sessions. Overall, our results demonstrate that NAC limits motivation, seeking behavior and reacquisition in rats, making it a potential new treatment for the maintenance of abstinence.
Collapse
Affiliation(s)
- Sophie Lebourgeois
- INSERM ERI-24 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé (CURS); Université de Picardie Jules Verne; France
| | - María Carmen González-Marín
- INSERM ERI-24 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé (CURS); Université de Picardie Jules Verne; France
| | - Jerome Jeanblanc
- INSERM ERI-24 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé (CURS); Université de Picardie Jules Verne; France
| | - Mickael Naassila
- INSERM ERI-24 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé (CURS); Université de Picardie Jules Verne; France
| | - Catherine Vilpoux
- INSERM ERI-24 GRAP, Groupe de Recherche sur l'Alcool et les Pharmacodépendances, Centre Universitaire de Recherche en Santé (CURS); Université de Picardie Jules Verne; France
| |
Collapse
|
89
|
Ruda-Kucerova J, Babinska Z, Luptak M, Getachew B, Tizabi Y. Both ketamine and NBQX attenuate alcohol drinking in male Wistar rats. Neurosci Lett 2018; 666:175-180. [PMID: 29288725 PMCID: PMC5805612 DOI: 10.1016/j.neulet.2017.12.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 11/18/2022]
Abstract
The devastating consequences of alcohol-use disorder (AUD) on the individual and the society are well established. Current treatments of AUD encompass various strategies, all of which have only modest effectiveness. Hence, there is a critical need to develop more efficacious therapies. Recently, specific glutamatergic receptors have been identified as potential novel targets for intervention in AUD. Thus, the current study was designed to evaluate the effects of acute administration of sub-anesthetic doses of ketamine, an NMDA receptor antagonist, as well as NBQX, an AMPA/kainate receptor antagonist on alcohol intake and its possible behavioural consequences. Adult male Wistar rats were trained in drinking in dark paradigm (3 weeks), and following stable alcohol intake, ketamine, NBQX as well as their combination were injected prior to a 90 min drinking session. In addition to alcohol intake, sucrose preference (overnight), and locomotor activity and forced swim test (FST) were also evaluated before and following alcohol intake. Both doses of ketamine (5 and 10 mg/kg) and NBQX (5 and 10 mg/kg) significantly attenuated percent alcohol intake. The combination of the higher dose of ketamine and NBQX, however, did not significantly affect percent alcohol intake. Moreover, animals exposed to alcohol showed decreased sucrose intake (reflective of anhedonia), decreased locomotor activity and swimming in the FST (reflective of helplessness), that were not affected by ketamine and/or NBQX. These results suggest that selective antagonism of the NMDA or AMPA/kainate receptors may be of therapeutic potential in AUD.
Collapse
Affiliation(s)
- Jana Ruda-Kucerova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Zuzana Babinska
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Matej Luptak
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
90
|
Kim R, Sepulveda-Orengo MT, Healey KL, Williams EA, Reissner KJ. Regulation of glutamate transporter 1 (GLT-1) gene expression by cocaine self-administration and withdrawal. Neuropharmacology 2018; 128:1-10. [PMID: 28919080 PMCID: PMC5714670 DOI: 10.1016/j.neuropharm.2017.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 12/24/2022]
Abstract
Downregulation of the astroglial glutamate transporter GLT-1 is observed in the nucleus accumbens (NAc) following administration of multiple drugs of abuse. The decrease in GLT-1 protein expression following cocaine self-administration is dependent on both the amount of cocaine self-administered and the length of withdrawal, with longer access to cocaine and longer withdrawal periods leading to greater decreases in GLT-1 protein. However, the mechanism(s) by which cocaine downregulates GLT-1 protein remains unknown. We used qRT-PCR to examine gene expression of GLT-1 splice isoforms (GLT-1A, GLT-1B) in the NAc, prelimbic cortex (PL) and basolateral amygdala (BLA) of rats, following two widely used models of cocaine self-administration: short-access (ShA) self-administration, and the long-access (LgA) self-administration/incubation model. While downregulation of GLT-1 protein is observed following ShA cocaine self-administration and extinction, this model did not lead to a change in GLT-1A or GLT-1B gene expression in any brain region examined. Forced abstinence following ShA cocaine self-administration also was without effect. In contrast, LgA cocaine self-administration and prolonged abstinence significantly decreased GLT-1A gene expression in the NAc and BLA, and significantly decreased GLT-1B gene expression in the PL. No change was observed in NAc GLT-1A gene expression one day after LgA cocaine self-administration, indicating withdrawal-induced decreases in GLT-1A mRNA. In addition, LgA cocaine self-administration and withdrawal induced hypermethylation of the GLT-1 gene in the NAc. These results indicate that a decrease in NAc GLT-1 mRNA is only observed after extended access to cocaine combined with protracted abstinence, and that epigenetic mechanisms likely contribute to this effect.
Collapse
Affiliation(s)
- Ronald Kim
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marian T Sepulveda-Orengo
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kati L Healey
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily A Williams
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn J Reissner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
91
|
A High-fat, High-sugar 'Western' Diet Alters Dorsal Striatal Glutamate, Opioid, and Dopamine Transmission in Mice. Neuroscience 2017; 372:1-15. [PMID: 29289718 DOI: 10.1016/j.neuroscience.2017.12.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Understanding neuroadaptations involved in obesity is critical for developing new approaches to treatment. Diet-induced neuroadaptations within the dorsal striatum have the capacity to drive excessive food seeking and consumption. Five-week-old C57BL/6J mice consumed a high-fat, high-sugar 'western diet' (WD) or a control 'standard diet' (SD) for 16 weeks. Weight gain, glucose tolerance, and insulin tolerance were measured to confirm an obese-like state. Following these 16 weeks, electrophysiological recordings were made from medium spiny neurons (MSNs) in the medial (DMS) and lateral (DLS) portions of dorsal striatum to evaluate diet effects on neuronal excitability and synaptic plasticity. In addition, fast-scan cyclic voltammetry evaluated dopamine transmission in these areas. WD mice gained significantly more weight and consumed more calories than SD mice and demonstrated impaired glucose tolerance. Electrophysiology data revealed that MSNs from WD mice demonstrated increased AMPA-to-NMDA receptor current ratio and prolonged spontaneous glutamate-mediated currents, specifically in the DLS. Evoked dopamine release was also significantly greater and reuptake slower in both subregions of WD striatum. Finally, dorsal striatal MSNs from WD mice were significantly less likely to demonstrate mu-opioid receptor-mediated synaptic plasticity. Neuronal excitability and GABAergic transmission were unaffected by diet in either striatal subregion. Our results demonstrate that a high-fat, high-sugar diet alters facets of glutamate, dopamine, and opioid signaling within the dorsal striatum, with some subregion specificity. These alterations within a brain area known to play a role in food motivation/consumption and habitual behavior are highly relevant for the clinical condition of obesity and its treatment.
Collapse
|
92
|
Adjunctive N-acetylcysteine in depression: exploration of interleukin-6, C-reactive protein and brain-derived neurotrophic factor. Acta Neuropsychiatr 2017; 29:337-346. [PMID: 28318471 DOI: 10.1017/neu.2017.2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This study aimed to explore effects of adjunctive N-acetylcysteine (NAC) treatment on inflammatory and neurogenesis markers in unipolar depression. METHODS We embarked on a 12-week clinical trial of NAC (2000 mg/day compared with placebo) as an adjunctive treatment for unipolar depression. A follow-up visit was conducted 4 weeks following the completion of treatment. We collected serum samples at baseline and the end of the treatment phase (week 12) to determine changes in interleukin-6 (IL6), C-reactive protein (CRP) and brain-derived neurotrophic factor (BDNF) following NAC treatment. RESULTS NAC treatment significantly improved depressive symptoms on the Montgomery-Asberg Depression Rating Scale (MADRS) over 16 weeks of the trial. Serum levels of IL6 were associated with reductions of MADRS scores independent of treatment response. However, we found no significant changes in IL6, CRP and BDNF levels following NAC treatment. CONCLUSION Overall, this suggests that our results failed to support the hypothesis that IL6, CRP and BDNF are directly involved in the therapeutic mechanism of NAC in depression. IL6 may be a useful marker for future exploration of treatment response.
Collapse
|
93
|
Krzyżanowska W, Pomierny B, Bystrowska B, Pomierny-Chamioło L, Filip M, Budziszewska B, Pera J. Ceftriaxone- and N-acetylcysteine-induced brain tolerance to ischemia: Influence on glutamate levels in focal cerebral ischemia. PLoS One 2017; 12:e0186243. [PMID: 29045497 PMCID: PMC5646803 DOI: 10.1371/journal.pone.0186243] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/27/2017] [Indexed: 01/22/2023] Open
Abstract
One of the major players in the pathophysiology of cerebral ischemia is disrupted homeostasis of glutamatergic neurotransmission, resulting in elevated extracellular glutamate (Glu) concentrations and excitotoxicity-related cell death. In the brain, Glu concentrations are regulated by Glu transporters, including Glu transporter-1 (GLT-1) and cystine/Glu antiporter (system xc-). Modulation of these transporters by administration of ceftriaxone (CEF, 200 mg/kg, i.p.) or N-acetylcysteine (NAC, 150 mg/kg, i.p.) for 5 days before focal cerebral ischemia may induce brain tolerance to ischemia by significantly limiting stroke-related damage and normalizing Glu concentrations. In the present study, focal cerebral ischemia was induced by 90-minute middle cerebral artery occlusion (MCAO). We compared the effects of CEF and NAC pretreatment on Glu concentrations in extracellular fluid and cellular-specific expression of GLT-1 and xCT with the effects of two reference preconditioning methods, namely, ischemic preconditioning and chemical preconditioning in rats. Both CEF and NAC significantly reduced Glu levels in the frontal cortex and hippocampus during focal cerebral ischemia, and this decrease was comparable with the Glu level achieved with the reference preconditioning strategies. The results of immunofluorescence staining of GLT-1 and xCT on astrocytes, neurons and microglia accounted for the observed changes in extracellular Glu levels to a certain extent. Briefly, after MCAO, the expression of GLT-1 on astrocytes decreased, but pretreatment with CEF seemed to prevent this downregulation. In addition, every intervention used in this study seemed to reduce xCT expression on astrocytes and neurons. The results of this study indicate that modulation of Glu transporter expression may restore Glu homeostasis. Moreover, our results suggest that CEF and NAC may induce brain tolerance to ischemia by influencing GLT-1 and system xc- expression levels. These transporters are presumably good targets for the development of novel therapies for brain ischemia.
Collapse
Affiliation(s)
- Weronika Krzyżanowska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Bartosz Pomierny
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
- * E-mail:
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Lucyna Pomierny-Chamioło
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Małgorzata Filip
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Drug Addiction Pharmacology, Kraków, Poland
| | - Bogusława Budziszewska
- Department of Biochemical Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
94
|
Rezvani AH, Levin ED, Cauley M, Getachew B, Tizabi Y. Ketamine Differentially Attenuates Alcohol Intake in Male Versus Female Alcohol Preferring (P) Rats. JOURNAL OF DRUG AND ALCOHOL RESEARCH 2017; 6:236030. [PMID: 29416900 PMCID: PMC5798244 DOI: 10.4303/jdar/236030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Although various pharmacological tools in combating addiction to alcohol are available, their efficacy is limited. Hence, there is a critical need for development of more effective medications. Recent advances in the field have identified the glutamatergic system as a potential novel target for intervention in addictive behaviors. PURPOSE Hence, we evaluated the effects of acute administration of low (subanesthetic) doses of ketamine, an NMDA receptor antagonist, on alcohol intake and alcohol preference in both male and female rats. STUDY DESIGN Adult alcohol preferring (P) rats were exposed to two-bottle choice (ethanol 10% and water) for at least three weeks following a nine-day training period and the effects of various doses of ketamine (5 mg/kg, 7.5 mg/kg, and 10 mg/kg, injected subcutaneously, SC) on consumption of alcohol over various time periods during a 24 h interval were measured. RESULTS Our results indicate that ketamine treatment significantly reduced both alcohol intake and preference in a time- and dose-dependent manner in both sexes. Moreover, a differential sensitivity between the sexes was observed. Thus, although alcohol intake was higher in males, female rats responded much more strongly to the highest dose of ketamine than males in the initial time periods. CONCLUSION It is concluded that glutamatergic receptor manipulations may be of therapeutic potential in addiction to alcohol and that different sexes may respond differentially to such treatments.
Collapse
Affiliation(s)
- Amir H. Rezvani
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Edward D. Levin
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Marty Cauley
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Bruk Getachew
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC 20059, USA
| | - Yousef Tizabi
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|
95
|
Shwartz A, Betzer O, Kronfeld N, Kazimirsky G, Cazacu S, Finniss S, Lee HK, Motiei M, Dagan SY, Popovtzer R, Brodie C, Yadid G. Therapeutic Effect of Astroglia-like Mesenchymal Stem Cells Expressing Glutamate Transporter in a Genetic Rat Model of Depression. Am J Cancer Res 2017; 7:2690-2703. [PMID: 28819456 PMCID: PMC5558562 DOI: 10.7150/thno.18914] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/15/2017] [Indexed: 12/12/2022] Open
Abstract
Recent studies have proposed that abnormal glutamatergic neurotransmission and glial pathology play an important role in the etiology and manifestation of depression. It was postulated that restoration of normal glutamatergic transmission, by enhancing glutamate uptake, may have a beneficial effect on depression. We examined this hypothesis using unique human glial-like mesenchymal stem cells (MSCs), which in addition to inherent properties of migration to regions of injury and secretion of neurotrophic factors, were differentiated to express high levels of functional glutamate transporters (excitatory amino acid transporters; EAAT). Additionally, gold nanoparticles (GNPs), which serve as contrast agents for CT imaging, were loaded into the cells for non-invasive, real-time imaging and tracking of MSC migration and final location within the brain. MSC-EAAT (2×105; 10 μl) were administered (i.c.v.) to Flinder Sensitive Line rats (FSLs), a genetic model for depression, and longitudinal behavioral and molecular changes were monitored. FSL rats treated with MSC-EAAT showed attenuated depressive-like behaviors (measured by the forced swim test, novelty exploration test and sucrose self-administration paradigm), as compared to controls. CT imaging, Flame Atomic Absorption Spectroscopy analysis and immunohistochemistry showed that the majority of MSCs homed specifically to the dentate gyrus of the hippocampus, a region showing structural brain changes in depression, including loss of glial cells. mRNA and protein levels of EAAT1 and BDNF were significantly elevated in the hippocampus of MSC-EAAT-treated FSLs. Our findings indicate that MSC-EAATs effectively improve depressive-like manifestations, possibly in part by increasing both glutamate uptake and neurotropic factor secretion in the hippocampus.
Collapse
|
96
|
Spencer S, Kalivas PW. Glutamate Transport: A New Bench to Bedside Mechanism for Treating Drug Abuse. Int J Neuropsychopharmacol 2017; 20:797-812. [PMID: 28605494 PMCID: PMC5632313 DOI: 10.1093/ijnp/pyx050] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/09/2017] [Indexed: 02/06/2023] Open
Abstract
Drug addiction has often been described as a "hijacking" of the brain circuits involved in learning and memory. Glutamate is the principal excitatory neurotransmitter in the brain, and its contribution to synaptic plasticity and learning processes is well established in animal models. Likewise, over the past 20 years the addiction field has ascribed a critical role for glutamatergic transmission in the development of addiction. Chronic drug use produces enduring neuroadaptations in corticostriatal projections that are believed to contribute to a maladaptive deficit in inhibitory control over behavior. Much of this research focuses on the role played by ionotropic glutamate receptors directly involved in long-term potentiation and depression or metabotropic receptors indirectly modulating synaptic plasticity. Importantly, the balance between glutamate release and clearance tightly regulates the patterned activation of these glutamate receptors, emphasizing an important role for glutamate transporters in maintaining extracellular glutamate levels. Five excitatory amino acid transporters participate in active glutamate reuptake. Recent evidence suggests that these glutamate transporters can be modulated by chronic drug use at a variety of levels. In this review, we synopsize the evidence and mechanisms associated with drug-induced dysregulation of glutamate transport. We then summarize the preclinical and clinical data suggesting that glutamate transporters offer an effective target for the treatment of drug addiction. In particular, we focus on the role that altered glutamate transporters have in causing drug cues and contexts to develop an intrusive quality that guides maladaptive drug seeking behaviors.
Collapse
Affiliation(s)
- Sade Spencer
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina.,Correspondence: Sade Spencer, PhD, Medical University of South Carolina, 173 Ashley Avenue, BSB, 403- MSC 510, Charleston, SC 29425 ()
| | - Peter W Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
97
|
Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, Roberts-Wolfe D, Kalivas PW. The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev 2017; 68:816-71. [PMID: 27363441 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - J A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - C D Gipson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - Y M Kupchik
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - S Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - A C W Smith
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - D Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| |
Collapse
|
98
|
Berridge KC, Robinson TE. Liking, wanting, and the incentive-sensitization theory of addiction. ACTA ACUST UNITED AC 2017; 71:670-679. [PMID: 27977239 DOI: 10.1037/amp0000059] [Citation(s) in RCA: 631] [Impact Index Per Article: 90.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rewards are both "liked" and "wanted," and those 2 words seem almost interchangeable. However, the brain circuitry that mediates the psychological process of "wanting" a particular reward is dissociable from circuitry that mediates the degree to which it is "liked." Incentive salience or "wanting," a form of motivation, is generated by large and robust neural systems that include mesolimbic dopamine. By comparison, "liking," or the actual pleasurable impact of reward consumption, is mediated by smaller and fragile neural systems, and is not dependent on dopamine. The incentive-sensitization theory posits the essence of drug addiction to be excessive amplification specifically of psychological "wanting," especially triggered by cues, without necessarily an amplification of "liking." This is because of long-lasting changes in dopamine-related motivation systems of susceptible individuals, called "neural sensitization." A quarter-century after its proposal, evidence has continued to grow in support the incentive-sensitization theory. Further, its scope is now expanding to include diverse behavioral addictions and other psychopathologies. (PsycINFO Database Record
Collapse
|
99
|
Minarini A, Ferrari S, Galletti M, Giambalvo N, Perrone D, Rioli G, Galeazzi GM. N-acetylcysteine in the treatment of psychiatric disorders: current status and future prospects. Expert Opin Drug Metab Toxicol 2016; 13:279-292. [PMID: 27766914 DOI: 10.1080/17425255.2017.1251580] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION N-acetylcysteine (NAC) is widely known for its role as a mucolytic and as an antidote to paracetamol overdose. There is increasing interest in the use of NAC in the treatment of several psychiatric disorders. The rationale for the administration of NAC in psychiatric conditions is based on its role as a precursor to the antioxidant glutathione, and its action as a modulating agent of glutamatergic, dopaminergic, neurotropic and inflammatory pathways. Areas covered: This study reviews the available data regarding the use of NAC in different psychiatric disorders including substance use disorders, autism, obsessive-compulsive spectrum disorders, schizophrenia, depression, bipolar disorder. Promising results were found in trials testing the use of NAC, mainly as an add-on treatment, in cannabis use disorder in young people, depression in bipolar disorder, negative symptoms in schizophrenia, and excoriation (skin-picking) disorder. Despite initial optimism, recent findings regarding NAC efficacy in autism have been disappointing. Expert opinion: These preliminary positive results require further confirmation in larger samples and with longer follow-ups. Given its high tolerability and wide availability, NAC represents an important target to investigate in the field of new adjunctive treatments for psychiatric conditions.
Collapse
Affiliation(s)
- Alessandro Minarini
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Silvia Ferrari
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Martina Galletti
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Nina Giambalvo
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Daniela Perrone
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Giulia Rioli
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| | - Gian Maria Galeazzi
- a Department of Diagnostic-Clinical Medicine and Public Health , University of Modena and Reggio Emilia , Modena , Italy
| |
Collapse
|
100
|
Israel Y, Ezquer F, Quintanilla ME, Morales P, Ezquer M, Herrera-Marschitz M. Intracerebral Stem Cell Administration Inhibits Relapse-like Alcohol Drinking in Rats. Alcohol Alcohol 2016; 52:1-4. [PMID: 27651282 DOI: 10.1093/alcalc/agw068] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022] Open
Abstract
Study describes the blockade of relapse-like alcohol drinking by mesenchymal stem cells (MSCs). High alcohol-intake bred rats consumed alcohol for 3 months and were subjected to repeated alcohol deprivations for 7-14 days, followed by alcohol reaccess. Upon reaccess, animals consumed 2.2 g alcohol/kg in 60 minutes. A single intra-cerebroventricular MSC administration inhibited relapse-like drinking up to 80-85% for 40 days (P < 0.001). An alcohol-use-disorder was prevented.
Collapse
Affiliation(s)
- Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|