1
|
Chakraborty S, Rao S, Tripathi SJ. The neuroprotective effects of N-acetylcysteine in psychiatric and neurodegenerative disorders: From modulation of glutamatergic transmission to restoration of synaptic plasticity. Neuropharmacology 2025:110527. [PMID: 40414419 DOI: 10.1016/j.neuropharm.2025.110527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 05/10/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
N-acetylcysteine (NAC) is an effective pleiotropic drug with a strong safety profile. It is predominantly used as a mucolytic agent and in the treatment of paracetamol overdose. However, extensive research in the last decade has shown the prominent efficacy of NAC in many neuropsychiatric and neurodegenerative disorders. NAC acts through multiple mechanisms; primarily, it releases cysteine and modulates glutamatergic and monoaminergic transmission. Further, it restores glutathione levels, promotes oxidative balance, reverses decreased synaptic plasticity, reduces neuroinflammation and mitochondrial dysfunction, and provides neurotrophic support. Additionally, it regulates one-carbon metabolism pathways, leading to the production of key metabolites. In this review, we will be discussing in-depth mechanisms of action of NAC and its promising ability to reverse neuropathological changes, particularly cognitive deficits, and associated plasticity changes in various psychiatric and neurodegenerative diseases, including depression, bipolar disorders, schizophrenia, Alzheimer's disease, Huntington's disease, traumatic brain injury, aging. Overall, several preclinical studies and clinical trials have demonstrated the efficacy of NAC in reversing regressive plasticity, cognitive deficits, and associated changes in the brain. NAC remains among the strongest candidates with a high safety profile for managing several types of neurological disorders.
Collapse
Affiliation(s)
- Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India.
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Bespalov A, Lütjens R, Doller D. Dusting off old blueprints: Is it time to reconsider metabotropic glutamate receptor 2 for therapeutic drug development? Pharmacol Biochem Behav 2025; 247:173908. [PMID: 39571688 DOI: 10.1016/j.pbb.2024.173908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025]
Abstract
The metabotropic glutamate receptor 2 (mGlu2) is a heavily studied therapeutic target in neuropsychiatry for which we anticipate a renewed interest in the near future. We review the rationale and the outcome of clinical trials with mGlu2/3 receptor agonists in schizophrenia, a field of intense research since a seminal publication by Patel and colleagues (2007). We summarize evidence about selective, potent and safe agents with quantifiable CNS penetration that can be used to test hypotheses of mGlu2 receptors involvement in neuropsychiatric diseases. We summarize lessons learned from previous programs that should be considered to maximize the probability of success when targeting orthosteric and allosteric enhancement of mGlu2 receptor function in schizophrenia and beyond. First, we propose expanding our focus beyond presynaptic mGlu2 receptor stimulation in schizophrenia to novel hypotheses and that the choice of a therapeutic indication no longer be dictated by commercial opportunity but following science as a driver. Second, evidence on internal validity of preclinical studies supporting efficacy claims in the mGlu2 field is very limited. This gap will need to be closed when reviewing the rationale to re-initiate efforts in this field. Third, the pomaglumetad program was halted due to insufficient clinical efficacy, partly because of the inability to identify a treatment responder population. In preclinical studies, effects of mGlu2/3 receptor stimulation also seemed to vary significantly between laboratories. Definition of the responsive subject population and development of response-predicting biomarkers is therefore one of the main avenues of further research in the mGlu2 field.
Collapse
|
3
|
Olaitan GO, Lynch WJ, Venton BJ. The therapeutic potential of low-intensity focused ultrasound for treating substance use disorder. Front Psychiatry 2024; 15:1466506. [PMID: 39628494 PMCID: PMC11612502 DOI: 10.3389/fpsyt.2024.1466506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/07/2024] [Indexed: 12/06/2024] Open
Abstract
Substance use disorder (SUD) is a persistent public health issue that necessitates the exploration of novel therapeutic interventions. Low-intensity focused ultrasound (LIFU) is a promising modality for precise and invasive modulation of brain activity, capable of redefining the landscape of SUD treatment. The review overviews effective LIFU neuromodulatory parameters and molecular mechanisms, focusing on the modulation of reward pathways in key brain regions in animal and human models. Integration of LIFU with established therapeutics holds promise for augmenting treatment outcomes in SUD. The current research examines LIFU's efficacy in reducing cravings and withdrawal symptoms. LIFU shows promise for reducing cravings, modulating reward circuitry, and addressing interoceptive dysregulation and emotional distress. Selecting optimal parameters, encompassing frequency, burst patterns, and intensity, is pivotal for balancing therapeutic efficacy and safety. However, inconsistencies in empirical findings warrant further research on optimal treatment parameters, physiological action mechanisms, and long-term effects. Collaborative interdisciplinary investigations are imperative to fully realize LIFU's potential in revolutionizing SUD treatment paradigms and enhancing patient outcomes.
Collapse
Affiliation(s)
- Greatness O. Olaitan
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Wendy J. Lynch
- Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, United States
| | - B. Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Littlepage-Saunders M, Hochstein MJ, Chang DS, Johnson KA. G protein-coupled receptor modulation of striatal dopamine transmission: Implications for psychoactive drug effects. Br J Pharmacol 2024; 181:4399-4413. [PMID: 37258878 PMCID: PMC10687321 DOI: 10.1111/bph.16151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Dopamine transmission in the striatum is a critical mediator of the rewarding and reinforcing effects of commonly misused psychoactive drugs. G protein-coupled receptors (GPCRs) that bind a variety of neuromodulators including dopamine, endocannabinoids, acetylcholine and endogenous opioid peptides regulate dopamine release by acting on several components of dopaminergic circuitry. Striatal dopamine release can be driven by both somatic action potential firing and local mechanisms that depend on acetylcholine released from striatal cholinergic interneurons. GPCRs that primarily regulate somatic firing of dopamine neurons via direct effects or modulation of synaptic inputs are likely to affect distinct aspects of behaviour and psychoactive drug actions compared with those GPCRs that primarily regulate local acetylcholine-dependent dopamine release in striatal regions. This review will highlight mechanisms by which GPCRs modulate dopaminergic transmission and the relevance of these findings to psychoactive drug effects on physiology and behaviour.
Collapse
Affiliation(s)
- Mydirah Littlepage-Saunders
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Michael J Hochstein
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Doris S Chang
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Illenberger JM, Flores-Ramirez FJ, Matzeu A, Lütjens R, Martin-Fardon R. ADX106772, an mGlu2 receptor positive allosteric modulator, selectively attenuates oxycodone taking and seeking. Neuropharmacology 2023; 238:109666. [PMID: 37463637 PMCID: PMC10529136 DOI: 10.1016/j.neuropharm.2023.109666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
Opioid abuse and overdose have risen to epidemic proportions in the United States. Oxycodone is the most abused prescription opioid. Treatments for opioid use disorder (OUD) seek to reduce vulnerability to relapse by reducing sources of reinforcement to seek drug (i.e., acute drug effects or drug withdrawal/craving). Accumulating evidence that glutamate release elicits drug-seeking behaviors has generated interest in pharmacotherapies targeting the glutamate system. Agonists and positive allosteric modulators of the metabotropic glutamate 2 (mGlu2) receptor decrease glutamate activity, reducing drug taking and seeking. The present study tested whether the mGlu2 receptor positive allosteric modulator ADX106772 reduces oxycodone self-administration and the conditioned reinstatement of oxycodone seeking without affecting behaviors directed toward a highly palatable nondrug reinforcer (sweetened condensed milk). Male Wistar rats were trained to self-administer oxycodone (0.15 mg/kg/infusion, i.v., 12 h/day) or sweetened condensed milk (SCM; diluted 2:1 v/v in H2O, orally, 30 min/day) for 13 days in the presence of a contextual/discriminative stimulus (SD), and the ability of ADX106772 (0, 0.3, 1, 3 and-10 mg/kg, s. c.) to decrease self-administration was tested. The rats then underwent extinction training, during which oxycodone, SCM, and the SD were withheld. After extinction, the ability of ADX106772 to prevent SD-induced conditioned reinstatement of oxycodone and SCM seeking was tested. ADX106772 reduced oxycodone self-administration and conditioned reinstatement without affecting SCM self-administration or conditioned reinstatement. ADX106772 reduced oxycodone taking and seeking and did not affect the motivation for the palatable conventional reinforcer, SCM, suggesting that activating mGlu2 receptors with a positive allosteric modulator is a potential approach for prescription OUD treatment.
Collapse
Affiliation(s)
- Jessica M Illenberger
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| | | | - Alessandra Matzeu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
6
|
Abstract
Diseases associated with nicotine dependence in the form of habitual tobacco use are a major cause of premature death in the United States. The majority of tobacco smokers will relapse within the first month of attempted abstinence. Smoking cessation agents increase the likelihood that smokers can achieve long-term abstinence. Nevertheless, currently available smoking cessation agents have limited utility and fail to prevent relapse in the majority of smokers. Pharmacotherapy is therefore an effective strategy to aid smoking cessation efforts but considerable risk of relapse persists even when the most efficacious medications currently available are used. The past decade has seen major breakthroughs in our understanding of the molecular, cellular, and systems-level actions of nicotine in the brain that contribute to the development and maintenance of habitual tobacco use. In parallel, large-scale human genetics studies have revealed allelic variants that influence vulnerability to tobacco use disorder. These advances have revealed targets for the development of novel smoking cessation agents. Here, we summarize current efforts to develop smoking cessation therapeutics and highlight opportunities for future efforts.
Collapse
Affiliation(s)
- Dana Lengel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul J. Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute (DDI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
7
|
Kong D, Sun JX, Yang JQ, Li YS, Bi K, Zhang ZY, Wang KH, Luo HY, Zhu M, Xu Y. Ketogenic diet: a potential adjunctive treatment for substance use disorders. Front Nutr 2023; 10:1191903. [PMID: 37575322 PMCID: PMC10414993 DOI: 10.3389/fnut.2023.1191903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Substance use disorders (SUD) can lead to serious health problems, and there is a great interest in developing new treatment methods to alleviate the impact of substance abuse. In recent years, the ketogenic diet (KD) has shown therapeutic benefits as a dietary therapy in a variety of neurological disorders. Recent studies suggest that KD can compensate for the glucose metabolism disorders caused by alcohol use disorder by increasing ketone metabolism, thereby reducing withdrawal symptoms and indicating the therapeutic potential of KD in SUD. Additionally, SUD often accompanies increased sugar intake, involving neural circuits and altered neuroplasticity similar to substance addiction, which may induce cross-sensitization and increased use of other abused substances. Reducing carbohydrate intake through KD may have a positive effect on this. Finally, SUD is often associated with mitochondrial damage, oxidative stress, inflammation, glia dysfunction, and gut microbial disorders, while KD may potentially reverse these abnormalities and serve a therapeutic role. Although there is much indirect evidence that KD has a positive effect on SUD, the small number of relevant studies and the fact that KD leads to side effects such as metabolic abnormalities, increased risk of malnutrition and gastrointestinal symptoms have led to the limitation of KD in the treatment of SUD. Here, we described the organismal disorders caused by SUD and the possible positive effects of KD, aiming to provide potential therapeutic directions for SUD.
Collapse
Affiliation(s)
- Deshenyue Kong
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-xue Sun
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji-qun Yang
- Third People’s Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, China
| | - Yuan-sen Li
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ke Bi
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zun-yue Zhang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Kun-hua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Hua-you Luo
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mei Zhu
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Xu
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Suchanecka A, Boroń A, Chmielowiec K, Strońska-Pluta A, Masiak J, Lachowicz M, Chmielowiec J, Grzywacz A. Association of the rs3864283 Polymorphism Located in the HINT1 Gene with Cigarette Use and Personality Traits. Int J Mol Sci 2023; 24:10244. [PMID: 37373392 PMCID: PMC10299401 DOI: 10.3390/ijms241210244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Nicotine is the major reinforcing component of tobacco and it is believed that the pharmacological effects of nicotine motivate the initiation and maintenance of a smoking habit. HINT1 appears to play a role in the modulation of the effects of drug abuse. Hence, the aim of this study was the analysis of the association between the rs3864283 polymorphism of the HINT1 gene and cigarette use; the analysis of personality traits assessed by the means of the NEO-FFI Inventory; the analysis of anxiety measured by the STAI questionnaire; and the analysis of the interactions between the rs3864283 and both personality traits and anxiety. The study group consisted of 522 volunteers. Of these, 371 were cigarette users and 151 were never-smokers. The genomic DNA was isolated from venous blood using standard procedures. The results of both inventories, i.e., NEO-FFI and STAI., were reported as the sten scores. Genotyping was conducted with the real-time PCR method. Statistically significant differences were found in the frequency of rs3864283 genotypes and alleles in the tested sample of Cigarette Users when compared to the control group. The Cigarette Users compared to the control group obtained higher scores in the assessment of NEO-FFI extraversion scale, and significantly lower results were obtained for the NEO-FFI openness scale, the agreeableness scale, and the conscientiousness scale. There was a statistically significant effect of rs3864283 genotype interaction and Cigarette Use or not using (control group) on the extraversion scale. There was also a statistically significant effect of Cigarette Users or the control group on the extraversion scale score. The results obtained in the presented study indicated a significant association between the HINT1 rs3864283 variant and smoking status. Moreover, this is the first study incorporating genetic association of above-mentioned polymorphic site with interaction analysis of personality traits and anxiety. Overall, the results of this study suggest that HINT1 is an important genetic component associated with nicotine usage mechanisms.
Collapse
Affiliation(s)
- Aleksandra Suchanecka
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| | - Agnieszka Boroń
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, Aleja Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland;
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (K.C.); (J.C.)
| | - Aleksandra Strońska-Pluta
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| | - Jolanta Masiak
- Second Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin, 1 Głuska St., 20-059 Lublin, Poland;
| | - Milena Lachowicz
- Department of Psychology, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
| | - Jolanta Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (K.C.); (J.C.)
| | - Anna Grzywacz
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| |
Collapse
|
9
|
Abarkan M, Fois GR, Vouillac-Mendoza C, Ahmed SH, Guillem K. Altered neuronal activity in the ventromedial prefrontal cortex drives nicotine intake escalation. Neuropsychopharmacology 2023; 48:887-896. [PMID: 36042320 PMCID: PMC10156690 DOI: 10.1038/s41386-022-01428-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/08/2022]
Abstract
Nicotine addiction develops after prolonged drug use and escalation of drug intake. However, because of difficulties in demonstrating escalation of nicotine use in rats, its underlying neuroadaptations still remain poorly understood. Here we report that access to unusually high doses of nicotine (i.e., from 30 µg to 240 µg/kg/injection) for self-administration precipitated a rapid and robust escalation of nicotine intake and increased the motivation for the drug in rats. This nicotine intake escalation also induced long-lasting changes in vmPFC neuronal activity both before and during nicotine self-administration. Specifically, after escalation of nicotine intake, basal vmPFC neuronal activity increased above pre-escalation and control activity levels, while ongoing nicotine self-administration restored these neuronal changes. Finally, simulation of the restoring effects of nicotine with in vivo optogenetic inhibition of vmPFC neurons caused a selective de-escalation of nicotine self-administration.
Collapse
Affiliation(s)
- Myriam Abarkan
- Université de Bordeaux, CNRS, Chimie et Biologie des Membranes et Nano-objets, UMR, 5248, Pessac, France
| | - Giulia R Fois
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France
| | | | - Serge H Ahmed
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France
| | - Karine Guillem
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000, Bordeaux, France.
| |
Collapse
|
10
|
Domi A, Lucente E, Cadeddu D, Adermark L. Nicotine but not saline self-administering or yoked control conditions produces sustained neuroadaptations in the accumbens shell. Front Mol Neurosci 2023; 16:1105388. [PMID: 36760603 PMCID: PMC9907443 DOI: 10.3389/fnmol.2023.1105388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Using yoked animals as the control when monitoring operant drug-self-administration is considered the golden standard. However, instrumental learning per se recruits several neurocircuits that may produce distinct or overlapping neuroadaptations with drugs of abuse. The aim of this project was to assess if contingent responding for nicotine or saline in the presence of a light stimulus as a conditioned reinforcer is associated with sustained neurophysiological adaptations in the nucleus accumbens shell (nAcS), a brain region repeatedly associated with reward related behaviors. Methods To this end, nicotine-or saline-administrating rats and yoked-saline stimulus-unpaired training conditions were assessed in operant boxes over four consecutive weeks. After four additional weeks of home cage forced abstinence and subsequent cue reinforced responding under extinction conditions, ex vivo electrophysiology was performed in the nAcS medium spiny neurons (MSNs). Results Whole cell recordings conducted in voltage and current-clamp mode showed that excitatory synapses in the nAcS were altered after prolonged forced abstinence from nicotine self-administration. We observed an increase in sEPSC amplitude in animals with a history of contingent nicotine SA potentially indicating higher excitability of accumbal MSNs, which was further supported by current clamp recordings. Interestingly no sustained neuroadaptations were elicited in saline exposed rats from nicotine associated visual cues compared to the yoked controls. Conclusion The data presented here indicate that nicotine self-administration produces sustained neuroadaptations in the nAcS while operant responding driven by nicotine visual stimuli has no long-term effects on MSNs in nAcS.
Collapse
Affiliation(s)
- Ana Domi
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,*Correspondence: Ana Domi, ✉
| | - Erika Lucente
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Davide Cadeddu
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise Adermark
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Preventing incubation of drug craving to treat drug relapse: from bench to bedside. Mol Psychiatry 2023; 28:1415-1429. [PMID: 36646901 DOI: 10.1038/s41380-023-01942-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/17/2023]
Abstract
In 1986, Gawin and Kleber reported a progressive increase in cue-induced drug craving in individuals with cocaine use disorders during prolonged abstinence. After years of controversy, as of 2001, this phenomenon was confirmed in rodent studies using self-administration model, and defined as the incubation of drug craving. The intensification of cue-induced drug craving after withdrawal exposes abstinent individuals to a high risk of relapse, which urged us to develop effective interventions to prevent incubated craving. Substantial achievements have been made in deciphering the neural mechanisms, with potential implications for reducing drug craving and preventing the relapse. The present review discusses promising drug targets that have been well investigated in animal studies, including some neurotransmitters, neuropeptides, neurotrophic factors, and epigenetic markers. We also discuss translational exploitation and challenges in the field of the incubation of drug craving, providing insights into future investigations and highlighting the potential of pharmacological interventions, environment-based interventions, and neuromodulation techniques.
Collapse
|
12
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
13
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|
14
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders. SIGNIFICANCE STATEMENT: Allosteric modulation of metabotropic glutamate (mGlu) receptors represents a promising therapeutic strategy to normalize dysregulated cellular physiology associated with neuropsychiatric disease. This review summarizes preclinical and clinical studies using mGlu receptor allosteric modulators as experimental tools and potential therapeutic approaches for the treatment of neuropsychiatric diseases, including schizophrenia, stress, and substance use disorders.
Collapse
|
15
|
Pham H, Seeley SL, D'Souza MS. Pharmacological activation of kappa opioid receptors in the nucleus accumbens core and ventral tegmental area increases the aversive effects of nicotine. Behav Pharmacol 2022; 33:266-281. [PMID: 35256559 DOI: 10.1097/fbp.0000000000000675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aversive effects of nicotine play an important role in the development of nicotine dependence. However, neural substrates and/or brain regions that play a role in the aversive effects of nicotine have not been fully identified. Previous work done in our laboratory showed that systemic administration of kappa opioid receptors (KORs) agonist ±U50488 increased the aversive effects of nicotine. In this study, we assessed the effects of KOR activation in specific brain regions, namely, the nucleus accumbens (NAcc) core and ventral tegmental area (VTA) on the aversive effects of nicotine using the conditioned taste aversion model. Separate groups of Wistar rats were implanted with cannulae above either the NAcc core or the VTA. KOR agonist (±U50488) was bilaterally infused in the NAcc core (0, 0.3, and 3 ug/0.5 ul/side) or VTA (0, 0.3, 1.5, and 3 ug/0.5 ul/side) prior to receiving nicotine (0.4 mg/kg, base; s.c.) during conditioning. Bilateral infusion of the KOR agonist (3 ug/0.5 ul/side) in the NAcc core or the VTA increased the aversive effects of nicotine compared with respective saline controls. Together, these results suggest that pharmacological activation of the KORs in the NAcc core and VTA dose dependently modulate the aversive effects of nicotine. Because aversive effects of nicotine determine susceptibility to development of nicotine dependence, we can conclude that KOR activity in the NAcc and VTA after administration of nicotine may determine susceptibility to the development of nicotine dependence.
Collapse
Affiliation(s)
- Han Pham
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, Ohio, USA
| | | | | |
Collapse
|
16
|
Abdullah M, Huang LC, Lin SH, Yang YK. Dopaminergic and glutamatergic biomarkers disruption in addiction and regulation by exercise: a mini review. Biomarkers 2022; 27:306-318. [PMID: 35236200 DOI: 10.1080/1354750x.2022.2049367] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Drug addiction is associated with disruption of a multitude of biomarkers in various brain regions, particularly in the reward center. The most pronounced are dopaminergic and glutamatergic biomarkers, which are affected at various levels. Neuropathological changes in biomarkers alter the homeostasis of the glutamatergic and dopaminergic nervous systems and promote addiction-associated characteristics such as repeated intake, maintenance, withdrawal, reinstatement, and relapse. Exercise has been shown to have a buffering effect on such biomarkers and reverse the effects of addictive substances. METHODS A review of the literature searched in PubMed, examining drug addiction and physical exercise in relation to dopaminergic and glutamatergic systems at any of the three biomarker levels (i.e., neurotransmitter, receptor, or transporter). RESULTS We review the collective impact of addictive substances on the dopaminergic and glutamatergic systems and the beneficial effect of exercise in terms of reversing the damage to these systems. We propose future directions, including implications of exercise as an add-on therapy, substance use disorder (SUD) prognosis and diagnosis and designing of optimized exercise and pharmaceutical regimens based on the aforementioned biomarkers. CONCLUSION Exercise is beneficial for all types of drug addiction at all stages, by reversing molecular damages caused to dopaminergic and glutamatergic systems.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan
| | - Li-Chung Huang
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, Chia-Yi Branch, Taichung Veterans General Hospital, Chia-Yi, Taiwan
| | - Shih-Hsien Lin
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Psychiatry, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| |
Collapse
|
17
|
Shipman ML, Corbit LH. Diet-induced deficits in goal-directed control are rescued by agonism of group II metabotropic glutamate receptors in the dorsomedial striatum. Transl Psychiatry 2022; 12:42. [PMID: 35091538 PMCID: PMC8799694 DOI: 10.1038/s41398-022-01807-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Many overweight or obese people struggle to sustain the behavioural changes necessary to achieve and maintain weight loss. In rodents, obesogenic diet can disrupt goal-directed control of responding for food reinforcers, which may indicate that diet can disrupt brain regions associated with behavioural control. We investigated a potential glutamatergic mechanism to return goal-directed control to rats who had been given an obesogenic diet prior to operant training. We found that an obesogenic diet reduced goal-directed control and that systemic injection of LY379268, a Group II metabotropic glutamate receptor (mGluR2/3) agonist, returned goal-directed responding in these rats. Further, we found that direct infusion of LY379268 into the dorsomedial striatum, a region associated with goal-directed control, also restored goal-directed responding in the obesogenic-diet group. This indicates that one mechanism through which obesogenic diet disrupts goal-directed control is glutamatergic, and infusion of a mGluR2/3 agonist into the DMS is sufficient to ameliorate deficits in goal-directed control.
Collapse
Affiliation(s)
- Megan L Shipman
- University of Toronto Department of Psychology, 100 St. George Street, Toronto, ON, M5S 3G3, Canada
| | - Laura H Corbit
- University of Toronto Department of Psychology, 100 St. George Street, Toronto, ON, M5S 3G3, Canada.
| |
Collapse
|
18
|
Stone TW. Relationships and Interactions between Ionotropic Glutamate Receptors and Nicotinic Receptors in the CNS. Neuroscience 2021; 468:321-365. [PMID: 34111447 DOI: 10.1016/j.neuroscience.2021.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
Although ionotropic glutamate receptors and nicotinic receptors for acetylcholine (ACh) have usually been studied separately, they are often co-localized and functionally inter-dependent. The objective of this review is to survey the evidence for interactions between the two receptor families and the mechanisms underlying them. These include the mutual regulation of subunit expression, which change the NMDA:AMPA response balance, and the existence of multi-functional receptor complexes which make it difficult to distinguish between individual receptor sites, especially in vivo. This is followed by analysis of the functional relationships between the receptors from work on transmitter release, cellular electrophysiology and aspects of behavior where these can contribute to understanding receptor interactions. It is clear that nicotinic receptors (nAChRs) on axonal terminals directly regulate the release of glutamate and other neurotransmitters, α7-nAChRs generally promoting release. Hence, α7-nAChR responses will be prevented not only by a nicotinic antagonist, but also by compounds blocking the indirectly activated glutamate receptors. This accounts for the apparent anticholinergic activity of some glutamate antagonists, including the endogenous antagonist kynurenic acid. The activation of presynaptic nAChRs is by the ambient levels of ACh released from pre-terminal synapses, varicosities and glial cells, acting as a 'volume neurotransmitter' on synaptic and extrasynaptic sites. In addition, ACh and glutamate are released as CNS co-transmitters, including 'cholinergic' synapses onto spinal Renshaw cells. It is concluded that ACh should be viewed primarily as a modulator of glutamatergic neurotransmission by regulating the release of glutamate presynaptically, and the location, subunit composition, subtype balance and sensitivity of glutamate receptors, and not primarily as a classical fast neurotransmitter. These conclusions and caveats should aid clarification of the sites of action of glutamate and nicotinic receptor ligands in the search for new centrally-acting drugs.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; Institute of Neuroscience, University of Glasgow, G12 8QQ, UK.
| |
Collapse
|
19
|
Nall RW, Heinsbroek JA, Nentwig TB, Kalivas PW, Bobadilla AC. Circuit selectivity in drug versus natural reward seeking behaviors. J Neurochem 2021; 157:1450-1472. [PMID: 33420731 PMCID: PMC8178159 DOI: 10.1111/jnc.15297] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Substance use disorder (SUD) is characterized, in part by behavior biased toward drug use and away from natural sources of reward (e.g., social interaction, food, sex). The neurobiological underpinnings of SUDs reveal distinct brain regions where neuronal activity is necessary for the manifestation of SUD-characteristic behaviors. Studies that specifically examine how these regions are involved in behaviors motivated by drug versus natural reward allow determinations of which regions are necessary for regulating seeking of both reward types, and appraisals of novel SUD therapies for off-target effects on behaviors motivated by natural reward. Here, we evaluate studies directly comparing regulatory roles for specific brain regions in drug versus natural reward. While it is clear that many regions drive behaviors motivated by all reward types, based on the literature reviewed we propose a set of interconnected regions that become necessary for behaviors motivated by drug, but not natural rewards. The circuitry is selectively necessary for drug seeking includes an Action/Reward subcircuit, comprising nucleus accumbens, ventral pallidum, and ventral tegmental area, a Prefrontal subcircuit comprising prelimbic, infralimbic, and insular cortices, a Stress subcircuit comprising the central nucleus of the amygdala and the bed nucleus of the stria terminalis, and a Diencephalon circuit including lateral hypothalamus. Evidence was mixed for nucleus accumbens shell, insular cortex, and ventral pallidum. Studies for all other brain nuclei reviewed supported a necessary role in regulating both drug and natural reward seeking. Finally, we discuss emerging strategies to further disambiguate the necessity of brain regions in drug- versus natural reward-associated behaviors.
Collapse
Affiliation(s)
- Rusty W. Nall
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- These authors share senior authorship
| | - Ana-Clara Bobadilla
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
- These authors share senior authorship
| |
Collapse
|
20
|
Aryal SP, Fu X, Sandin JN, Neupane KR, Lakes JE, Grady ME, Richards CI. Nicotine induces morphological and functional changes in astrocytes via nicotinic receptor activity. Glia 2021; 69:2037-2053. [PMID: 33851731 DOI: 10.1002/glia.24011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 01/16/2023]
Abstract
Nicotine is a highly addictive compound present in tobacco, which causes the release of dopamine in different regions of the brain. Recent studies have shown that astrocytes express nicotinic acetylcholine receptors (nAChRs) and mediate calcium signaling. In this study, we examine the morphological and functional adaptations of astrocytes due to nicotine exposure. Utilizing a combination of fluorescence and atomic force microscopy, we show that nicotine-treated astrocytes exhibit time-dependent remodeling in the number and length of both proximal and fine processes. Blocking nAChR activity with an antagonist completely abolishes nicotine's influence on astrocyte morphology indicating that nicotine's action is mediated by these receptors. Functional studies show that 24-hr nicotine treatment induces higher levels of calcium activity in both the cell soma and the processes with a more substantial change observed in the processes. Nicotine does not induce reactive astrocytosis even at high concentrations (10 μM) as determined by cytokine release and glial fibrillary acidic protein expression. We designed tissue clearing experiments to test whether morphological changes occur in vivo using astrocyte specific Aldh1l1-tdTomato knock in mice. We find that nicotine induces a change in the volume of astrocytes in the prefrontal cortex, CA1 of the hippocampus, and the substantia nigra. These results indicate that nicotine directly alters the functional and morphological properties of astrocytes potentially contributing to the underlying mechanism of nicotine abuse.
Collapse
Affiliation(s)
- Surya P Aryal
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Xu Fu
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Joree N Sandin
- Department of Mechanical Engineering, University of Kentucky, Lexington, Kentucky, USA
| | - Khaga R Neupane
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Jourdan E Lakes
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky, Lexington, Kentucky, USA
| | | |
Collapse
|
21
|
Yang JH, Sohn S, Kim S, Kim J, Oh JH, Ryu IS, Go BS, Choe ES. Repeated nicotine exposure increases the intracellular interaction between ERK-mGluR5 in the nucleus accumbens more in adult than adolescent rats. Addict Biol 2021; 26:e12913. [PMID: 32339332 DOI: 10.1111/adb.12913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/20/2020] [Accepted: 04/09/2020] [Indexed: 11/27/2022]
Abstract
Intracellular interactions between protein kinases and metabotropic receptors in the striatum regulate behavioral changes in response to drug exposure. We investigated the difference in the degree of interaction between extracellular signal-regulated kinase (ERK) and metabotropic glutamate receptor subtype 5 (mGluR5) in the nucleus accumbens (NAc) after repeated exposure to nicotine in adult and adolescent rats. The results showed that repeated exposure to nicotine (0.5 mg/kg/day, s.c.) for seven consecutive days increased ERK phosphorylation more in adults than in adolescents. Furthermore, membrane expression of mGluR5 in gamma-aminobutyric acid (GABA) medium spiny neurons was higher in adults than adolescents as a result of repeated exposure to nicotine. Blockade of mGluR5 with MPEP (0.5 nmol/side) decreased the repeated nicotine-induced increase in ERK phosphorylation. Either blockade of mGluR5 or inhibition of ERK with SL327 (150 nmol/side) decreased the repeated nicotine-induced increase in the level of inositol-1,4,5-triphosphate (IP3 ), a key transducer associated with mGluR5-coupled signaling cascades. Similarly, interference of binding between activated ERK and mGluR5 by the blocking peptide, Tat-mGluR5-i (2 nmol/side), decreased the repeated nicotine-induced increases in IP3 and locomotor activity in adults. These findings suggest that the intracellular interaction between ERK and mGluR5 in the NAc is stronger in adult than in adolescent rats, which enhances the understanding of age-associated behavioral changes that occur after repeated exposure to nicotine.
Collapse
Affiliation(s)
- Ju Hwan Yang
- Department of Biological Sciences Pusan National University Busan South Korea
| | - Sumin Sohn
- Department of Biological Sciences Pusan National University Busan South Korea
| | - Sunghyun Kim
- Department of Biological Sciences Pusan National University Busan South Korea
| | - Jieun Kim
- Department of Biological Sciences Pusan National University Busan South Korea
| | - Jeong Hwan Oh
- Department of Biological Sciences Pusan National University Busan South Korea
- Institute of Fisheries Sciences Pukyong National University Busan South Korea
| | - In Soo Ryu
- Department of Biological Sciences Pusan National University Busan South Korea
- Korea Institute of Toxicology Daejeon South Korea
| | - Bok Soon Go
- Department of Biological Sciences Pusan National University Busan South Korea
| | - Eun Sang Choe
- Department of Biological Sciences Pusan National University Busan South Korea
| |
Collapse
|
22
|
Machado RCBR, Vargas HO, Baracat MM, Urbano MR, Verri WA, Porcu M, Nunes SOV. N-acetylcysteine as an adjunctive treatment for smoking cessation: a randomized clinical trial. ACTA ACUST UNITED AC 2020; 42:519-526. [PMID: 32725102 PMCID: PMC7524413 DOI: 10.1590/1516-4446-2019-0753] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/21/2020] [Indexed: 02/05/2023]
Abstract
Objective: This randomized controlled trial examined the efficacy and safety of N-acetylcysteine as an adjunctive treatment for smoking cessation. Methods: Heavy smokers were recruited from smoking cessation treatment for this 12- week randomized controlled trial. Eligible tobacco use disorder outpatients (n=34) were randomized to N-acetylcysteine or placebo plus first-line treatment. Abstinence was verified by exhaled carbon monoxide (COexh). The assessment scales included the Fagerström Test for Nicotine Dependence, the Hamilton Depression Rating Scale, the Hamilton Anxiety Rating Scale, the Minnesota Nicotine Withdrawal Scale, and the Medication Adherence Rating Scale. We also assessed anthropometrics, blood pressure, lipid profile, and soluble tumor necrosis factor receptor (sTNF-R) levels 1 and 2. Results: First-line treatment for smoking cessation plus adjunctive N-acetylcysteine or placebo significantly reduced COexh (p < 0.01). In the N-acetylcysteine group, no significant changes were found in nicotine withdrawal symptoms, depressive and anxiety symptoms, anthropometric measures, blood pressure, or glucose compared to placebo. However, there was a significant reduction in sTNF-R2 levels between baseline and week 12 in the N-acetylcysteine group. Conclusions: These findings highlight the need to associate N-acetylcysteine with first-line treatment for smoking cessation, since combined treatment may affect inflammation and metabolism components. Clinical trial registration: NCT02420418
Collapse
Affiliation(s)
- Regina C B R Machado
- Centro de Referência de Abordagem e Tratamento do Tabagismo, Hospital Universitário, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil
| | - Heber O Vargas
- Centro de Referência de Abordagem e Tratamento do Tabagismo, Hospital Universitário, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil.,Departamento de Medicina Clínica, Unidade de Psiquiatria, Hospital Universitário, CCS, UEL, Londrina, PR, Brazil
| | - Marcela M Baracat
- Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil
| | - Mariana R Urbano
- Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil.,Departamento de Estatística, Centro de Ciências Exatas, UEL, Londrina, PR, Brazil
| | - Waldiceu A Verri
- Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil.,Departamento de Patologia, Centro de Ciências Biológicas, UEL, Londrina, PR, Brazil
| | - Mauro Porcu
- Centro de Referência de Abordagem e Tratamento do Tabagismo, Hospital Universitário, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil
| | - Sandra O V Nunes
- Centro de Referência de Abordagem e Tratamento do Tabagismo, Hospital Universitário, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Centro de Ciências da Saúde (CCS), UEL, Londrina, PR, Brazil.,Departamento de Medicina Clínica, Unidade de Psiquiatria, Hospital Universitário, CCS, UEL, Londrina, PR, Brazil
| |
Collapse
|
23
|
Effects of 3-Month Exposure to E-Cigarette Aerosols on Glutamatergic Receptors and Transporters in Mesolimbic Brain Regions of Female C57BL/6 Mice. TOXICS 2020; 8:toxics8040095. [PMID: 33137879 PMCID: PMC7712012 DOI: 10.3390/toxics8040095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022]
Abstract
Electronic cigarettes (e-cigs) use has been dramatically increased recently, especially among youths. Previous studies from our laboratory showed that chronic exposure to e-cigs, containing 24 mg/mL nicotine, was associated with dysregulation of glutamate transporters and neurotransmitter levels in the brain of a mouse model. In this study, we evaluated the effect of three months’ continuous exposure to e-cig vapor (JUUL pods), containing a high nicotine concentration, on the expression of glutamate receptors and transporters in drug reward brain regions such as the nucleus accumbens (NAc) core (NAc-core), NAc shell (NAc-shell) and hippocampus (HIP) in female C57BL/6 mice. Three months’ exposure to mint- or mango-flavored JUUL (containing 5% nicotine, 59 mg/mL) induced upregulation of metabotropic glutamate receptor 1 (mGluR1) and postsynaptic density protein 95 (phosphorylated and total PSD95) expression, and downregulation of mGluR5 and glutamate transporter 1 (GLT-1) in the NAc-shell. In addition, three months’ exposure to JUUL was associated with upregulation of mGluR5 and GLT-1 expression in the HIP. These findings demonstrated that three-month exposure to e-cig vapor containing high nicotine concentrations induced differential effects on the glutamatergic system in the NAc and HIP, suggesting dysregulation of glutamatergic system activity in mesolimbic brain regions.
Collapse
|
24
|
Short-term nicotine deprivation alters dorsal anterior cingulate glutamate concentration and concomitant cingulate-cortical functional connectivity. Neuropsychopharmacology 2020; 45:1920-1930. [PMID: 32559759 PMCID: PMC7608204 DOI: 10.1038/s41386-020-0741-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/20/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Most cigarette smokers who wish to quit too often relapse within the first few days of abstinence, primarily due to the aversive aspects of the nicotine withdrawal syndrome (NWS), which remains poorly understood. Considerable research has suggested that the dorsal anterior cingulate cortex (dACC) plays a key role in nicotine dependence, with its functional connections between other brain regions altered as a function of trait addiction and state withdrawal. The flow of information between dACC and fronto-striatal regions is secured through different pathways, the vast majority of which are glutamatergic. As such, we investigated dACC activity using resting state functional connectivity (rsFC) with functional magnetic resonance imaging (fMRI) and glutamate (Glu) concentration with magnetic resonance spectroscopy (MRS). We also investigated the changes in adenosine levels in plasma during withdrawal as a surrogate for brain adenosine, which plays a role in fine-tuning synaptic glutamate transmission. Using a double-blind, placebo-controlled, randomized crossover design, nontreatment seeking smoking participants (N = 30) completed two imaging sessions, one while nicotine sated and another after 36 h nicotine abstinence. We observed reduced dACC Glu (P = 0.029) along with a significant reduction in plasma adenosine (P = 0.03) and adenosine monophosphate (AMP; P < 0.0001) concentrations during nicotine withdrawal in comparison with nicotine sated state. This withdrawal state manipulation also led to an increase in rsFC strength (P < 0.05) between dACC and several frontal cortical regions, including left superior frontal gyrus (LSFG), and right middle frontal gyrus (RMFG). Moreover, the state-trait changes in dACC Glu and rsFC strength between the dACC and both SFG and MFG were positively correlated (P = 0.012, and P = 0.007, respectively). Finally, the change in circuit strength between dACC and LSFG was negatively correlated with the change in withdrawal symptom manifestations as measured by the Wisconsin Smoking Withdrawal Scale (P = 0.04) and Tobacco Craving Questionnaire (P = 0.014). These multimodal imaging-behavioral findings reveal the complex cascade of changes induced by acute nicotine deprivation and call for further investigation into the potential utility of adenosine- and glutamate-signaling as novel therapeutic targets to treat the NWS.
Collapse
|
25
|
Johnson KA, Voyvodic L, Loewinger GC, Mateo Y, Lovinger DM. Operant self-stimulation of thalamic terminals in the dorsomedial striatum is constrained by metabotropic glutamate receptor 2. Neuropsychopharmacology 2020; 45:1454-1462. [PMID: 31995814 PMCID: PMC7360544 DOI: 10.1038/s41386-020-0626-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 12/25/2022]
Abstract
Dorsal striatal manipulations including stimulation of dopamine release and activation of medium spiny neurons (MSNs) are sufficient to drive reinforcement-based learning. Glutamatergic innervation of the striatum by the cortex and thalamus is a critical determinant of MSN activity and local regulation of dopamine release. However, the relationship between striatal glutamatergic afferents and behavioral reinforcement is not well understood. We evaluated the reinforcing properties of optogenetic stimulation of thalamostriatal terminals, which are associated with vesicular glutamate transporter 2 (Vglut2) expression, in the dorsomedial striatum (DMS), a region implicated in goal-directed behaviors. In mice expressing channelrhodopsin-2 (ChR2) under control of the Vglut2 promoter, optical stimulation of the DMS reinforced operant lever-pressing behavior. Mice also acquired operant self-stimulation of thalamostriatal terminals when ChR2 expression was virally targeted to the intralaminar thalamus. Stimulation trains that supported operant responding evoked dopamine release in the DMS and excitatory postsynaptic currents in DMS MSNs. Our previous work demonstrated that the presynaptic G protein-coupled receptor metabotropic glutamate receptor 2 (mGlu2) robustly inhibits glutamate and dopamine release induced by activation of thalamostriatal afferents. Thus, we examined the regulation of thalamostriatal self-stimulation by mGlu2. Administration of an mGlu2/3 agonist or an mGlu2-selective positive allosteric modulator reduced self-stimulation. Conversely, blockade of these receptors increased thalamostriatal self-stimulation, suggesting that endogenous activation of these receptors negatively modulates the reinforcing properties of thalamostriatal activity. These findings demonstrate that stimulation of thalamic terminals in the DMS is sufficient to reinforce a self-initiated action, and that thalamostriatal reinforcement is constrained by mGlu2 activation.
Collapse
Affiliation(s)
- Kari A. Johnson
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA ,0000 0001 0421 5525grid.265436.0Present Address: Department of Pharmacology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, C2019, Bethesda, MD 20814 USA
| | - Lucas Voyvodic
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| | - Gabriel C. Loewinger
- 000000041936754Xgrid.38142.3cDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - Yolanda Mateo
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| | - David M. Lovinger
- 0000 0001 2297 5165grid.94365.3dLaboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD 20852 USA
| |
Collapse
|
26
|
Moro F, Giannotti G, Caffino L, Marzo CM, Di Clemente A, Fumagalli F, Cervo L. Lasting reduction of nicotine-seeking behavior by chronic N-acetylcysteine during experimental cue-exposure therapy. Addict Biol 2020; 25:e12771. [PMID: 31132808 DOI: 10.1111/adb.12771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 02/10/2019] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
Nicotine-associated cues can trigger reinstatement in humans as well as in animal models of drug addiction. To date, no behavioral intervention or pharmacological treatment has been effective in preventing relapse in the long term. A large body of evidence indicates that N-acetylcysteine (N-AC) blunts the activation of glutamatergic (GLUergic) neurons in the nucleus accumbens (Nacc) associated with reinstatement. We evaluated the effect of an experimental cue exposure therapy (eCET) alone or in combination with N-AC to verify whether restoring GLU homeostasis enhances extinction of nicotine-associated cues. Rats were trained to associate discriminative stimuli with intravenous nicotine or saline self-administration. Reinforced response was followed by cue signals. After rats met the self-administration criteria, the lasting anti-relapse activity of i.p. N-AC or vehicle was assessed in three different experimental conditions after 14 days of treatment: treatment + eCET; treatment + lever-presses extinction (LP-EXT); and treatment + abstinence. N-AC 100 mg/kg, but not 60 mg/kg, induced anti-relapse activity that persisted 50 days after treatment only when paired with either LP-EXT or eCET with the greater activity found in the latter condition. To identify potential mechanisms for behavioral results, separate groups of rats that received either N-AC or vehicle + eCET were killed at different time points for Nacc Western-blot analysis. Seven days after treatment, chronic N-AC restored the expression of proteins crucial for GLU homeostasis, while at 50 days, it increased the expression of type II metabotropic GLU receptors. These results suggest that N-AC treatment in combination with eCET may offer a novel strategy to prevent relapse in nicotine addiction.
Collapse
Affiliation(s)
- Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| | | | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | | | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Milan Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research—IRCCS Milan Italy
| |
Collapse
|
27
|
Giacometti LL, Barker JM. Sex differences in the glutamate system: Implications for addiction. Neurosci Biobehav Rev 2020; 113:157-168. [PMID: 32173404 DOI: 10.1016/j.neubiorev.2020.03.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 01/21/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022]
Abstract
Clinical and preclinical research have identified sex differences in substance use and addiction-related behaviors. Historically, substance use disorders are more prevalent in men than women, though this gap is closing. Despite this difference, women appear to be more susceptible to the effects of many drugs and progress to substance abuse treatment more quickly than men. While the glutamate system is a key regulator of addiction-related behaviors, much of the work implicating glutamate signaling and glutamatergic circuits has been conducted in men and male rodents. An increasing number of studies have identified sex differences in drug-induced glutamate alterations as well as sex and estrous cycle differences in drug seeking behaviors. This review will describe sex differences in the glutamate system with an emphasis on implications for substance use disorders, highlighting the gaps in our current understanding of how innate and drug-induced alterations in the glutamate system may contribute to sex differences in addiction-related behaviors.
Collapse
Affiliation(s)
- L L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| | - J M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, United States.
| |
Collapse
|
28
|
Johnson KA, Lovinger DM. Allosteric modulation of metabotropic glutamate receptors in alcohol use disorder: Insights from preclinical investigations. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:193-232. [PMID: 32416868 DOI: 10.1016/bs.apha.2020.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are family C G protein-coupled receptors (GPCRs) that modulate neuronal excitability and synaptic transmission throughout the nervous system. Owing to recent advances in development of subtype-selective allosteric modulators of mGlu receptors, individual members of the mGlu receptor family have been proposed as targets for treating a variety of neurological and psychiatric disorders, including substance use disorders. In this chapter, we highlight preclinical evidence that allosteric modulators of mGlu receptors could be useful for reducing alcohol consumption and preventing relapse in alcohol use disorder (AUD). We begin with an overview of the preclinical models that are used to study mGlu receptor involvement in alcohol-related behaviors. Alcohol exposure causes adaptations in both expression and function of various mGlu receptor subtypes, and pharmacotherapies aimed at reversing these adaptations have the potential to reduce alcohol consumption and seeking. Positive allosteric modulators (PAMs) of mGlu2 and negative allosteric modulators of mGlu5 show particular promise for reducing alcohol intake and/or preventing relapse. Finally, this chapter discusses important considerations for translating preclinical findings toward the development of clinically useful drugs, including the potential for PAMs to avoid tolerance issues that are frequently observed with repeated administration of GPCR agonists.
Collapse
Affiliation(s)
- Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
29
|
Waeiss RA, Knight CP, Engleman EA, Hauser SR, Rodd ZA. Co-administration of ethanol and nicotine heightens sensitivity to ethanol reward within the nucleus accumbens (NAc) shell and increasing NAc shell BDNF is sufficient to enhance ethanol reward in naïve Wistar rats. J Neurochem 2020; 152:556-569. [PMID: 31721205 PMCID: PMC10826843 DOI: 10.1111/jnc.14914] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/26/2022]
Abstract
Alcohol use disorder most commonly presents as a polydrug disorder where greater than 85% are estimated to smoke. EtOH and nicotine (NIC) co-abuse or exposure results in unique neuroadaptations that are linked to behaviors that promote drug use. The current experiments aimed to identify neuroadaptations within the mesolimbic pathway produced by concurrent EtOH and NIC exposure. The experiments used four overall groups of male Wistar rats consisting of vehicle, EtOH or NIC alone, and EtOH+NIC. Drug exposure through direct infusion into the posterior ventral tegmental area (pVTA) stimulated release of glutamate and dopamine in the nucleus accumbens (NAc) shell, which was quantified through high-performance liquid chromatography. Additionally, brain-derived neurotrophic factor (BDNF) protein levels were measured via enzyme-linked immunosorbent assay (ELISA). A second experiment investigated the effects of drug pretreatment within the pVTA on the reinforcing properties of EtOH within the NAc shell through intracranial self-administration (ICSA). The concluding experiment evaluated the effect of NAc shell pretreatment with BDNF on EtOH reward utilizing ICSA within that region. The data indicated that only EtOH+NIC administration into the pVTA simultaneously increased glutamate, dopamine, and BDNF in the NAc shell. Moreover, only pVTA pretreatment with EtOH+NIC enhanced the reinforcing properties of EtOH in the NAc shell. BDNF pretreatment in the NAc shell was also sufficient to enhance the reinforcing properties of EtOH in the NAc shell. The collected data suggest that concurrent EtOH+NIC exposure results in a distinct neurochemical response and neuroadaptations within the mesolimbic pathway that alter EtOH reward.
Collapse
Affiliation(s)
- Robert A Waeiss
- Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher P Knight
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Eric A Engleman
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheketha R Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zachary A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Gendy MNS, Ibrahim C, Sloan ME, Le Foll B. Randomized Clinical Trials Investigating Innovative Interventions for Smoking Cessation in the Last Decade. Handb Exp Pharmacol 2020; 258:395-420. [PMID: 31267165 DOI: 10.1007/164_2019_253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Every year, billions of dollars are spent treating smoking and related conditions, yet smoking-related morbidity and mortality continue to rise. There are currently only three FDA-approved medications for smoking cessation: nicotine replacement therapy, bupropion, and varenicline. Although these medications increase abstinence rates, most individuals relapse following treatment. This chapter reviews clinical trials published within the past 10 years investigating novel smoking cessation pharmacotherapies. Among these pharmacotherapies, some showed promising results, such as cytisine and endocannabinoid modulators, whereas others failed to produce significant effects. More research is needed to develop drugs that produce higher rates of long-term abstinence and to determine which subgroups of patients benefit from a given treatment.
Collapse
Affiliation(s)
- Marie N S Gendy
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Christine Ibrahim
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Matthew E Sloan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada.
- Addictions Division, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Alcohol Research and Treatment Clinic, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Thorpe HHA, Hamidullah S, Jenkins BW, Khokhar JY. Adolescent neurodevelopment and substance use: Receptor expression and behavioral consequences. Pharmacol Ther 2019; 206:107431. [PMID: 31706976 DOI: 10.1016/j.pharmthera.2019.107431] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Adolescence is the transitional period between childhood and adulthood, during which extensive brain development occurs. Since this period also overlaps with the initiation of drug use, it is important to consider how substance use during this time might produce long-term neurobiological alterations, especially against the backdrop of developmental changes in neurotransmission. Alcohol, cannabis, nicotine, and opioids all produce marked changes in the expression and function of the neurotransmitter and receptor systems with which they interact. These acute and chronic alterations also contribute to behavioral consequences ranging from increased addiction risk to cognitive or neuropsychiatric behavioral dysfunctions. The current review provides an in-depth overview and update of the developmental changes in neurotransmission during adolescence, as well as the impact of drug exposure during this neurodevelopmental window. While most of these factors have been studied in animal models, which are the focus of this review, future longitudinal studies in humans that assess neural function and behavior will help to confirm pre-clinical findings. Furthermore, the neural changes induced by each drug should also be considered in the context of other contributing factors, such as sex. Further understanding of these consequences can help in the identification of novel approaches for preventing and reversing the neurobiological effects of adolescent substance use.
Collapse
Affiliation(s)
- Hayley H A Thorpe
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Shahnaza Hamidullah
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Bryan W Jenkins
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Jibran Y Khokhar
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada.
| |
Collapse
|
32
|
Activation of PPARγ Attenuates the Expression of Physical and Affective Nicotine Withdrawal Symptoms through Mechanisms Involving Amygdala and Hippocampus Neurotransmission. J Neurosci 2019; 39:9864-9875. [PMID: 31685649 DOI: 10.1523/jneurosci.1922-19.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022] Open
Abstract
An isoform of peroxisome proliferator-activated receptors (PPARs), PPARγ, is the receptor for the thiazolidinedione class of anti-diabetic medications including pioglitazone. Neuroanatomical data indicate PPARγ localization in brain areas involved in drug addiction. Preclinical and clinical data have shown that pioglitazone reduces alcohol and opioid self-administration, relapse to drug seeking, and plays a role in emotional responses. Here, we investigated the behavioral effect of PPARγ manipulation on nicotine withdrawal in male Wistar rats and in male mice with neuron-specific PPARγ deletion (PPARγ(-/-)) and their littermate wild-type (PPARγ(+/+)) controls. Real-time quantitative RT-PCR and RNAscope in situ hybridization assays were used for assessing the levels of expression and cell-type localization of PPARγ during nicotine withdrawal. Brain site-specific microinjections of the PPARγ agonist pioglitazone were performed to explore the role of this system on nicotine withdrawal at a neurocircuitry level. Results showed that activation of PPARγ by pioglitazone abolished the expression of somatic and affective nicotine withdrawal signs in rats and in (PPARγ(+/+)) mice. This effect was blocked by the PPARγ antagonist GW9662. During early withdrawal and protracted abstinence, the expression of PPARγ increased in GABAergic and glutamatergic cells of the amygdala and hippocampus, respectively. Hippocampal microinjections of pioglitazone reduced the expression of the physical signs of withdrawal, whereas excessive anxiety associated with protracted abstinence was prevented by pioglitazone microinjection into the amygdala. Our results demonstrate the implication of the neuronal PPARγ in nicotine withdrawal and indicates that activation of PPARγ may offer an interesting strategy for smoking cessation.SIGNIFICANCE STATEMENT Smoking cessation leads the occurrence of physical and affective withdrawal symptoms representing a major burden to quit tobacco use. Here, we show that activation of PPARγ prevents the expression of both somatic and affective signs of nicotine withdrawal. At molecular levels results show that PPARγ expression increases in GABAergic cells in the hippocampus and in GABA- and glutamate-positive cells in the basolateral amygdala. Hippocampal microinjections of pioglitazone reduce the insurgence of the physical withdrawal signs, whereas anxiety linked to protracted abstinence is attenuated by pioglitazone injected into the amygdala. Our results demonstrate the implication of neuronal PPARγ in nicotine withdrawal and suggest that PPARγ agonism may represent a promising treatment to aid smoking cessation.
Collapse
|
33
|
Lainiola M, Hietala L, Linden AM, Aitta-Aho T. The lack of conditioned place preference, but unaltered stimulatory and ataxic effects of alcohol in mGluR3-KO mice. J Psychopharmacol 2019; 33:855-864. [PMID: 31070489 DOI: 10.1177/0269881119844178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Alcohol use associates with environmental cues that can later reinstate drinking patterns without any alcohol exposure. Alcohol-induced reward, when combined with contextual signals of various sensory modalities in the central synapses of mesolimbic reward circuitries, can lead to the formation of conditioned responses. AIMS As the activation of glutamatergic synapses is pivotal in such processes, we aimed to investigate whether the metabotropic glutamate receptor subtype 3 plays a role in alcohol-induced behaviours including place preference. METHODS The metabotropic glutamate receptor subtype 3 knockout (mGluR3-KO) mouse line was used to study alcohol-induced place preference, locomotor activating and ataxic effects, limited access alcohol drinking, and preference for sucrose and saccharin. RESULTS Alcohol-induced horizontal locomotor stimulation and reduced rearing behaviour remained unchanged in the mGluR3-KO mice. However, alcohol-induced place conditioning in an unbiased paradigm setup was lacking in the mGluR3-KO mice, but clearly present in wildtype mice. Locomotor activity was not different between the mGluR3-KO and wildtype mice during the acquisition and expression trials. Alcohol consumption, studied through the 'drinking in the dark' model, remained unchanged in the mGluR3-KO mice, although low consumption in both wildtype and knockout mice hampers interpretation. The mGluR3-KO mice also showed normal sucrose and saccharin preference. CONCLUSIONS These studies indicate a role for metabotropic glutamate receptor subtype 3 in the conditioned contextual alcohol responses, but not in stimulatory, and ataxic alcohol effects.
Collapse
Affiliation(s)
- Mira Lainiola
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Lana Hietala
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Anni-Maija Linden
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Schmidt HD, Rupprecht LE, Addy NA. Neurobiological and Neurophysiological Mechanisms Underlying Nicotine Seeking and Smoking Relapse. MOLECULAR NEUROPSYCHIATRY 2019; 4:169-189. [PMID: 30815453 PMCID: PMC6388439 DOI: 10.1159/000494799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022]
Abstract
Tobacco-related morbidity and mortality continue to be a significant public health concern. Unfortunately, current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, a better understanding of the neurobiological and neurophysiological mechanisms that promote smoking relapse is needed to develop novel smoking cessation medications. Here, we review preclinical studies focused on identifying the neurotransmitter and neuromodulator systems that mediate nicotine relapse, often modeled in laboratory animals using the reinstatement paradigm, as well as the plasticity-dependent neurophysiological mechanisms that facilitate nicotine reinstatement. Particular emphasis is placed on how these neuroadaptations relate to smoking relapse in humans. We also highlight a number of important gaps in our understanding of the neural mechanisms underlying nicotine reinstatement and critical future directions, which may lead toward the development of novel, target pharmacotherapies for smoking cessation.
Collapse
Affiliation(s)
- Heath D. Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E. Rupprecht
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
- Interdepartmental Neuroscience Program, Yale Graduate School of Arts and Sciences, New Haven, Connecticut, USA
| |
Collapse
|
35
|
Microinjection of the mGluR2/3 agonist, LY379268, into the nucleus accumbens attenuates extinction latencies and the reinstatement of morphine-induced conditioned place preference in rats. Behav Pharmacol 2018; 29:385-392. [DOI: 10.1097/fbp.0000000000000375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Caprioli D, Justinova Z, Venniro M, Shaham Y. Effect of Novel Allosteric Modulators of Metabotropic Glutamate Receptors on Drug Self-administration and Relapse: A Review of Preclinical Studies and Their Clinical Implications. Biol Psychiatry 2018; 84:180-192. [PMID: 29102027 PMCID: PMC5837933 DOI: 10.1016/j.biopsych.2017.08.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022]
Abstract
Results from preclinical rodent studies during the last 20 years implicated glutamate neurotransmission in different brain regions in drug self-administration and rodent models of relapse. These results, along with evidence for drug-induced neuroadaptations in glutamatergic neurons and receptors, suggested that addiction might be treatable by medications that inhibit glutamatergic responses to drugs of abuse, drug-associated cues, and stressors. This idea is supported by findings in rodent and primate models that drug self-administration and relapse are reduced by systemic injections of antagonists of ionotropic glutamate receptors or metabotropic glutamate receptors (mGluRs) or orthosteric agonists of mGluR2/3. However, these compounds have not advanced to clinical use because of potential side effects and other factors. This state of affairs has led to the development of positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) of mGluRs. PAMs and NAMs of mGluRs, either of which can inhibit evoked glutamate release, may be suitable for testing in humans. We reviewed results from recent studies of systemically injected PAMs and NAMs of mGluRs in rodents and monkeys, focusing on whether they reduce drug self-administration, reinstatement of drug seeking, and incubation of drug craving. We also review results from rat studies in which PAMs or NAMs of mGluRs were injected intracranially to reduce drug self-administration and reinstatement. We conclude that PAMs and NAMs of mGluRs should be considered for clinical trials.
Collapse
Affiliation(s)
- Daniele Caprioli
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy.
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Marco Venniro
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| |
Collapse
|
37
|
Abstract
INTRODUCTION Tobacco use causes one premature death every six seconds. Current smoking cessation aids include nicotine replacement therapies, bupropion, and varenicline. Although more than 70% of smokers express a desire to quit, fewer than 3% remain abstinent for more than one year, highlighting a critical need for more efficacious smoking cessation treatments. Areas covered: The authors discuss the rationale, preclinical and clinical development of varenicline for smoking cessation. They cover the development of varenicline as a partial agonist at α4β2 receptors, the primary neural substrate for nicotine reward. Then, they discuss evidence from preclinical studies indicating varenicline's efficacy in blocking nicotine reward, followed by clinical trials demonstrating safety and efficacy in sustaining abstinence in smokers. Finally, they cover post-market surveillance, including caution in heavy machine operators, putative cardiovascular risk, and the repealed warning for adverse neuropsychiatric events. Expert opinion: Varenicline development was based on strong theoretical rationale and preclinical evidence. Clinical studies indicate that varenicline is safe and more effective in sustaining abstinence than placebo, bupropion or nicotine replacement therapies. However, given that continuous abstinence rates across studies remain low (18 ~ 30% with varenicline; 4 ~ 10% with placebo), novel and more effective medications targeting other nicotinic or glutamate receptors for smoking cessation are required.
Collapse
Affiliation(s)
- Chloe J. Jordan
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
38
|
Kenny PJ, Hoyer D, Koob GF. Animal Models of Addiction and Neuropsychiatric Disorders and Their Role in Drug Discovery: Honoring the Legacy of Athina Markou. Biol Psychiatry 2018; 83:940-946. [PMID: 29602521 DOI: 10.1016/j.biopsych.2018.02.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/28/2022]
Abstract
Each of the co-authors worked with Athina Markou, at different stages of our careers and in different capacities, to develop, optimize, and use animal models of drug addiction and, more generally, mental health disorders such as anxiety, depression, and schizophrenia. Here, we briefly summarize some of our work with Athina, primarily involving the use of the intracranial self-stimulation and intravenous drug self-administration procedures. This work established that excessive consumption of addictive drugs can induce profound dysfunction in brain reward circuits. Such drug-induced reward deficits are likely to play a key role in precipitating the emergence of compulsive drug-seeking behaviors. We also summarize findings suggesting that perturbations in glutamatergic transmission contribute to brain reward deficits in drug-dependent animals and that metabotropic glutamate receptors are potential targets for the development of novel medications to facilitate long-term drug abstinence and prevention of relapse.
Collapse
Affiliation(s)
- Paul J Kenny
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California; and the National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland
| |
Collapse
|
39
|
Cross AJ, Anthenelli R, Li X. Metabotropic Glutamate Receptors 2 and 3 as Targets for Treating Nicotine Addiction. Biol Psychiatry 2018; 83:947-954. [PMID: 29301614 PMCID: PMC5953779 DOI: 10.1016/j.biopsych.2017.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
Abstract
Tobacco smoking, driven by the addictive properties of nicotine, continues to be a worldwide health problem. Based on the well-established role of glutamatergic neurotransmission in drug addiction, novel medication development strategies seek to halt nicotine consumption and prevent relapse to tobacco smoking by modulating glutamate transmission. The presynaptic inhibitory metabotropic glutamate receptors 2 and 3 (mGluR2/3) are key autoreceptors on glutamatergic terminals that maintain glutamate homeostasis. Accumulating evidence suggests the critical role of mGluR2/3 in different aspects of nicotine addiction, including acquisition and maintenance of nicotine taking, nicotine withdrawal, and persistent nicotine seeking even after prolonged abstinence. The involvement of mGluR2/3 in other neuropsychiatric conditions, such as anxiety, depression, schizophrenia, Alzheimer's disease, Parkinson's disease, and pain, provides convincing evidence suggesting that mGluR2/3 may provide an effective therapeutic approach for comorbidity of smoking and these conditions. This focused review article highlights that mGluR2/3 provide a promising target in the search for smoking cessation medication with novel mechanisms of actions that differ from those of currently U.S. Food and Drug Administration-approved pharmacotherapies.
Collapse
Affiliation(s)
- Alan J Cross
- AstraZeneca Neuroscience Innovative Medicines, Cambridge, Massachusetts
| | - Robert Anthenelli
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California
| | - Xia Li
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
40
|
Namba MD, Tomek SE, Olive MF, Beckmann JS, Gipson CD. The Winding Road to Relapse: Forging a New Understanding of Cue-Induced Reinstatement Models and Their Associated Neural Mechanisms. Front Behav Neurosci 2018; 12:17. [PMID: 29479311 PMCID: PMC5811475 DOI: 10.3389/fnbeh.2018.00017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
In drug addiction, cues previously associated with drug use can produce craving and frequently trigger the resumption of drug taking in individuals vulnerable to relapse. Environmental stimuli associated with drugs or natural reinforcers can become reliably conditioned to increase behavior that was previously reinforced. In preclinical models of addiction, these cues enhance both drug self-administration and reinstatement of drug seeking. In this review, we will dissociate the roles of conditioned stimuli as reinforcers from their modulatory or discriminative functions in producing drug-seeking behavior. As well, we will examine possible differences in neurobiological encoding underlying these functional differences. Specifically, we will discuss how models of drug addiction and relapse should more systematically evaluate these different types of stimuli to better understand the neurobiology underlying craving and relapse. In this way, behavioral and pharmacotherapeutic interventions may be better tailored to promote drug use cessation outcomes and long-term abstinence.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Seven E. Tomek
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Joshua S. Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Cassandra D. Gipson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
41
|
Windisch KA, Czachowski CL. Effects of group II metabotropic glutamate receptor modulation on ethanol- and sucrose-seeking and consumption in the rat. Alcohol 2018; 66:77-85. [PMID: 29220747 DOI: 10.1016/j.alcohol.2017.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 11/30/2022]
Abstract
Previous studies suggest that group II metabotropic glutamate receptors (mGluR2/3) are involved in regulating ethanol-seeking and consumption. The mGluR2/3 agonist LY379268 (LY37) and selective mGluR2 positive allosteric modulator biphenyl‑indanone A (BINA) were used to investigate the relative contribution of mGlu2 and mGlu3 receptors on ethanol- and sucrose-seeking and consumption. A microinjection study was then performed to examine the role of nucleus accumbens (NAc) core mGluR2/3 on ethanol-seeking. For the systemic experiments, separate groups of male Wistar rats [LY37 (0-2.0 mg/kg); BINA (0-20 mg/kg)] were trained to complete a response requirement (RR) resulting in access to 10% ethanol or 2% sucrose (in separate groups) for a 20‑min drinking period. Animals then underwent consummatory testing (weekly drug injections with RR1) followed by appetitive testing (weekly drug injections followed by extinction session). A separate group of male Wistar rats was surgically implanted with bilateral guide cannulae directed toward the NAc core and had weekly microinjections followed by an extinction session. Systemic administration of the mGluR2/3 agonist LY37 significantly reduced ethanol- and sucrose-seeking. The same treatment also reduced sucrose consumption and body weight (24‑h post injection). Systemic administration of the selective mGluR2 PAM BINA, however, had no effect on either seeking or consumption of ethanol or sucrose. Intra-accumbens core LY37 significantly reduced ethanol-seeking. These findings suggest that systemic mGluR2/3 agonism, but not allosteric modulation of mGluR2, reduces reinforcer-seeking. In particular, NAc core group II mGluR may be involved in regulating ethanol-seeking.
Collapse
Affiliation(s)
- Kyle A Windisch
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| | - Cristine L Czachowski
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
42
|
Markou A, Li J, Tse K, Li X. Cue-induced nicotine-seeking behavior after withdrawal with or without extinction in rats. Addict Biol 2018; 23:111-119. [PMID: 27894160 DOI: 10.1111/adb.12480] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 11/28/2022]
Abstract
Exposure to smoking-associated environmental cues during smoke cessation elicits self-reported urge/craving to smoke, which precipitates relapse even after prolonged abstinence. Incubation of cue-induced cigarettes craving during abstinence has been observed in human smokers recently. The present studies assessed cue-induced nicotine-seeking behavior under different withdrawal conditions in rats with a history of nicotine self-administration. We found that non-reinforced operant responding during cue-induced nicotine seeking after different periods of withdrawal from nicotine exhibited an inverted U-shaped curve, with higher levels of responding after 7-21 days of withdrawal than those after 1-day withdrawal. Cue-induced nicotine-seeking responding is long lasting and persists even after 42 days of forced withdrawal in the home cages. Interestingly, repeated testing of cue-induced nicotine seeking at different withdrawal time points (1, 7, 14, 21 and 42 days) in the same individual alleviated responding as compared with the between-subjects assessment. Furthermore, extinction training during nicotine withdrawal significantly decreased cue-induced reinstatement of nicotine-seeking behavior. Together, profound time-dependent incubation of cue-induced craving in nicotine-experienced rats were observed. In addition, repeated cue exposure or extinction training decreases cue-induced craving. The demonstration of incubation of nicotine craving phenomenon in both rat and human studies provides support for the translational potential of therapeutic targets for relapse uncovered through mechanism studies in rats.
Collapse
Affiliation(s)
- Athina Markou
- Department of Psychiatry, School of Medicine; University of California San Diego; La Jolla CA USA
| | - Jie Li
- Department of Psychiatry, School of Medicine; University of California San Diego; La Jolla CA USA
| | - Kearny Tse
- Department of Psychiatry, School of Medicine; University of California San Diego; La Jolla CA USA
| | - Xia Li
- Department of Psychiatry, School of Medicine; University of California San Diego; La Jolla CA USA
| |
Collapse
|
43
|
Klenowski PM, Tapper AR. Molecular, Neuronal, and Behavioral Effects of Ethanol and Nicotine Interactions. Handb Exp Pharmacol 2018; 248:187-212. [PMID: 29423839 DOI: 10.1007/164_2017_89] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
44
|
Moro F, Orrù A, Marzo CM, Di Clemente A, Cervo L. mGluR2/3 mediates short-term control of nicotine-seeking by acute systemic N-acetylcysteine. Addict Biol 2018; 23:28-40. [PMID: 27558879 DOI: 10.1111/adb.12443] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/24/2016] [Accepted: 08/01/2016] [Indexed: 12/26/2022]
Abstract
Chronic self-administration of nicotine induces maladaptive changes in the cortico-accumbal glutamate (Glu) network. Consequently, re-exposure to nicotine-associated cues raises extracellular Glu in the nucleus accumbens reinstating drug-seeking. Restoring basal concentrations of extracellular Glu, thereby increasing tonic activation of the presynaptic group II metabotropic Glu receptors (mGluR2/3) with N-acetylcysteine (N-AC), might offer a valid therapeutic approach for maintaining smoking abstinence. Although N-AC modulates nicotine-seeking behavior by drug-associated stimuli in abstinent rats, it is still unclear whether it occurs through activation of mGluR2/3. Male Wistar rats were trained to associate discriminative stimuli (SD s) with the availability of intravenous nicotine (0.03 mg/kg/65 µl/2-second/infusion) or oral saccharin (100 µl of 50 mg/l) self-administration versus non-reward. Reinforced response was followed by a cue signaling 20-second time-out (CSs). Once the training criterion was met, rats underwent lever press extinction, without reinforcers, SD s and CSs. Re-exposure to nicotine or saccharin SD+ /CS+ , but not non-reward SD- /CS- , revived responding on the previously reinforced lever. Acute N-AC, 100 but not 60 or 30 mg/kg i.p., reduced cue-induced nicotine-seeking. N-AC 100 mg/kg did not modify cue-induced saccharin-seeking behavior or influenced locomotor activity. Blocking mGluR2/3 with the selective antagonist LY341495, 1 mg/kg i.p., completely prevented the antirelapse activity of N-AC. The finding that N-AC prevents cue-induced nicotine-seeking by stimulating mGluR2/3 might indicate a therapeutic opportunity for acute cue-controlled nicotine-seeking. Future studies could evaluate the persistent effects of chronic N-AC in promoting enduring suppression of nicotine-cue conditioned responding.
Collapse
Affiliation(s)
- Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Alessandro Orrù
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Claudio Marcello Marzo
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience; IRCCS-Mario Negri Institute for Pharmacological Research; Italy
| |
Collapse
|
45
|
Behavioral changes after nicotine challenge are associated with α7 nicotinic acetylcholine receptor-stimulated glutamate release in the rat dorsal striatum. Sci Rep 2017; 7:15009. [PMID: 29118361 PMCID: PMC5678080 DOI: 10.1038/s41598-017-15161-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/23/2017] [Indexed: 01/28/2023] Open
Abstract
Neurochemical alterations associated with behavioral responses induced by re-exposure to nicotine have not been sufficiently characterized in the dorsal striatum. Herein, we report on changes in glutamate concentrations in the rat dorsal striatum associated with behavioral alterations after nicotine challenge. Nicotine challenge (0.4 mg/kg/day, subcutaneous) significantly increased extracellular glutamate concentrations up to the level observed with repeated nicotine administration. This increase occurred in parallel with an increase in behavioral changes in locomotor and rearing activities. In contrast, acute nicotine administration and nicotine withdrawal on days 1 and 6 did not alter glutamate levels or behavioral changes. Blockade of α7 nicotinic acetylcholine receptors (nAChRs) significantly decreased the nicotine challenge-induced increases in extracellular glutamate concentrations and locomotor and rearing activities. These findings suggest that behavioral changes in locomotor and rearing activities after re-exposure to nicotine are closely associated with hyperactivation of the glutamate response by stimulating α7 nAChRs in the rat dorsal striatum.
Collapse
|
46
|
Duailibi MS, Cordeiro Q, Brietzke E, Ribeiro M, LaRowe S, Berk M, Trevizol AP. N-acetylcysteine in the treatment of craving in substance use disorders: Systematic review and meta-analysis. Am J Addict 2017; 26:660-666. [DOI: 10.1111/ajad.12620] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/12/2017] [Accepted: 08/17/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
| | - Quirino Cordeiro
- Department of Psychiatry; Federal University of São Paulo; São Paulo Brazil
| | - Elisa Brietzke
- Department of Psychiatry; Federal University of São Paulo; São Paulo Brazil
| | - Marcelo Ribeiro
- Department of Psychiatry; Federal University of São Paulo; São Paulo Brazil
- Reference Center for Alcohol, Tobacco and Other Drugs (CRATOD); São Paulo State Secretariat of Health; São Paulo São Paulo Brazil
| | - Steve LaRowe
- Department of Psychiatry; Center for Drug and Alcohol Programs; Medical University of South Carolina; Charleston South Carolina
| | - Michael Berk
- School of Medicine; IMPACT Strategic Research Centre; Deakin University; Geelong Victoria Australia
- Department of Psychiatry; The Florey Institute of Neuroscience and Mental Health, and Orygen Youth Health Research Centre; University of Melbourne; Parkville Victoria Australia
| | - Alisson Paulino Trevizol
- Reference Center for Alcohol, Tobacco and Other Drugs (CRATOD); São Paulo State Secretariat of Health; São Paulo São Paulo Brazil
| |
Collapse
|
47
|
Miranda MI, Rangel-Hernández JA, Vera-Rivera G, García-Medina NE, Soto-Alonso G, Rodríguez-García G, Núñez-Jaramillo L. The role of dopamine D2 receptors in the nucleus accumbens during taste-aversive learning and memory extinction after long-term sugar consumption. Neuroscience 2017; 359:142-150. [DOI: 10.1016/j.neuroscience.2017.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 10/19/2022]
|
48
|
Hicks C, Gregg RA, Nayak SU, Cannella LA, Schena GJ, Tallarida CS, Reitz AB, Smith GR, Rawls SM. Glutamate carboxypeptidase II (GCPII) inhibitor 2-PMPA reduces rewarding effects of the synthetic cathinone MDPV in rats: a role for N-acetylaspartylglutamate (NAAG). Psychopharmacology (Berl) 2017; 234:1671-1681. [PMID: 28251297 PMCID: PMC5433920 DOI: 10.1007/s00213-017-4568-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 02/13/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE Metabotropic glutamate 2 and 3 (mGluR2/3) receptors are implicated in drug addiction as they limit excessive glutamate release during relapse. N-acetylaspartylglutamate (NAAG) is an endogenous mGluR2/3 agonist that is inactivated by the glutamate carboxypeptidase II (GCPII) enzyme. GCPII inhibitors, and NAAG itself, attenuate cocaine-seeking behaviors. However, their effects on the synthetic cathinone 3,4-methylenedioxypyrovalerone (MDPV) have not been examined. OBJECTIVES We determined whether withdrawal following repeated MDPV administration alters GCPII expression in corticolimbic regions. We also examined whether a GCPII inhibitor (2-(phosphonomethyl)-pentanedioic acid (2-PMPA)), and NAAG, reduce the rewarding and locomotor-stimulant effects of MDPV in rats. METHODS GCPII was assessed following repeated MDPV exposure (7 days). The effects of 2-PMPA and NAAG on acute MDPV-induced hyperactivity were determined using a locomotor test. We also examined the inhibitory effects of 2-PMPA and NAAG on MDPV-induced place preference, and whether the mGluR2/3 antagonist LY341495 could prevent these effects. RESULTS MDPV withdrawal reduced GCPII expression in the prefrontal cortex. Systemic injection of 2-PMPA (100 mg/kg) did not affect the hyperactivity produced by MDPV (0.5-3 mg/kg). However, nasal administration of NAAG did reduce MDPV-induced ambulation, but only at the highest dose (500 μg/10 μl). We also showed that 2-PMPA (10-30 mg/kg) and NAAG (10-500 μg/10 μl) dose-dependently attenuated MDPV place preference, and that the effect of NAAG was blocked by LY341495 (3 mg/kg). CONCLUSIONS These findings demonstrate that MDPV withdrawal produces dysregulation in the endogenous NAAG-GCPII signaling pathway in corticolimbic circuitry. Systemic administration of the GCPII inhibitor 2-PMPA, or NAAG, attenuates MDPV reward.
Collapse
Affiliation(s)
- Callum Hicks
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ryan A Gregg
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Sunil U Nayak
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Lee Anne Cannella
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Giana J Schena
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Christopher S Tallarida
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Doylestown, PA, USA
| | - Garry R Smith
- Fox Chase Chemical Diversity Center, Doylestown, PA, USA
| | - Scott M Rawls
- Department of Pharmacology, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
49
|
KK-92A, a novel GABA B receptor positive allosteric modulator, attenuates nicotine self-administration and cue-induced nicotine seeking in rats. Psychopharmacology (Berl) 2017; 234:1633-1644. [PMID: 28382542 DOI: 10.1007/s00213-017-4594-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/11/2017] [Indexed: 01/03/2023]
Abstract
RATIONALE GABAB receptors (GABABR) play a critical role in GABAergic neurotransmission in the brain and are thought to be one of the most promising targets for the treatment of drug addiction. GABABR positive allosteric modulators (PAMs) have shown promise as potential anti-addictive therapies, as they lack the sedative and muscle relaxant properties of full GABAB receptor agonists such as baclofen. OBJECTIVES The present study was aimed at developing novel, selective, and potent GABABR PAMs with efficacy on abuse-related effects of nicotine. RESULTS We synthetized ~100 analogs of BHF177, a GABABR PAM that has been shown to inhibit nicotine taking and seeking, and tested their activity in multiple cell-based functional assays. Among these compounds, KK-92A displayed superior PAM properties at the GABABR. Interestingly, our results revealed the existence of pathway-selective differential modulation of GABABR signaling by the structurally related GABABR allosteric modulators BHF177 and KK-92A. In vivo, similarly to BHF177, KK-92A inhibited intravenous nicotine self-administration under both fixed- and progressive-ratio schedules of reinforcement in rats. In contrast to BHF177, KK-92A had no effect on food self-administration. Furthermore, KK-92A decreased cue-induced nicotine-seeking behavior without affecting food seeking. CONCLUSIONS These results indicate that KK-92A is a selective GABABR PAM with efficacy in inhibition of the primary reinforcing and incentive motivational effects of nicotine, and attenuation of nicotine seeking, further confirming that GABABR PAMs may be useful antismoking medications.
Collapse
|
50
|
Sturchler E, Li X, de Lourdes Ladino M, Kaczanowska K, Cameron M, Griffin PR, Finn MG, Markou A, McDonald P. GABA B receptor allosteric modulators exhibit pathway-dependent and species-selective activity. Pharmacol Res Perspect 2017; 5:e00288. [PMID: 28357120 PMCID: PMC5368958 DOI: 10.1002/prp2.288] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/02/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Positive modulation of the GABAB receptor (GABABR) represents a potentially useful therapeutic approach for the treatment of nicotine addiction. The positive allosteric modulators (PAMs) of GABABR GS39783 and BHF177 enhance GABA‐stimulated [35S]GTPγS‐binding, and have shown efficacy in a rodent nicotine self‐administration procedure reflecting aspects of nicotine dependence. Interestingly, the structural related analog, NVP998, had no effect on nicotine self‐administration in rats despite demonstrating similar pharmacokinetic properties. Extensive in vitro characterization of GS39783, BHF177, and NVP998 activity on GABABR‐regulated signaling events, including modulation of cAMP, intracellular calcium levels, and ERK activation, revealed that these structurally related molecules display distinct pathway‐specific signaling activities that correlate with the dissimilarities observed in rodent models and may be predictive of in vivo efficacy. Furthermore, these GABABR allosteric modulators exhibit species‐dependent activity. Collectively, these data will be useful in guiding the development of GABABR allosteric modulators that display optimal in vivo efficacy in a preclinical model of nicotine dependence, and will identify those that have the potential to lead to novel antismoking therapies.
Collapse
Affiliation(s)
- Emmanuel Sturchler
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Xia Li
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Maria de Lourdes Ladino
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458; Present address: School of Medicine Vanderbilt University 2215 Garland Ave Nashville Tennessee 37232
| | - Kasia Kaczanowska
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Department of Chemistry University of California San Diego, 9500 Gilman Drive La Jolla California 92093
| | - Michael Cameron
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - Patrick R Griffin
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| | - M G Finn
- Department of Chemistry The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037; Present address: Georgia Institute of Technology School of Chemistry and Biochemistry 901 Atlantic Drive Atlanta Georgia 30332
| | - Athina Markou
- Department of Psychiatry University of California San Diego 9500 Gilman Drive La Jolla California 92093
| | - Patricia McDonald
- Department of Molecular Therapeutics The Scripps Research Institute 130 Scripps way Jupiter Florida 33458
| |
Collapse
|