1
|
Ballard IC, Pappas I, Furman DJ, Berry AS, deB Frederick B, White RL, Kayser AS, Jagust WJ, D'Esposito M. Temporal fMRI Dynamics Map Dopamine Physiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645022. [PMID: 40196490 PMCID: PMC11974834 DOI: 10.1101/2025.03.24.645022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Spatial variations in dopamine function are linked to cognition and substance use disorders but are challenging to characterize with current methods. Because dopamine influences blood vessel dilation, we hypothesized that hemodynamic latency, which reflects BOLD signal timing, could serve as an indirect marker of dopamine physiology. Across four datasets, we found a topography of hemodynamic latencies that precisely distinguished the nucleus accumbens, a dopaminergic region implicated in motivation and substance abuse, from other striatal regions. Using PET, genetics, and pharmacology, we found that hemodynamic latencies are robustly related to dopamine function and dopamine-linked behavior. In individuals with cocaine use disorder, we observed a spatial gradient of altered hemodynamic latencies in the striatum. This pattern independently predicted nicotine use, revealing a conserved physiological profile associated with addictive substance use. Hemodynamic latencies map regional, individual, and pathological differences linked to dopamine, opening new avenues for indirectly assessing the role of dopamine in healthy cognition and disease.
Collapse
Affiliation(s)
- Ian C Ballard
- Psychology Department, University of California, Riverside
| | - Ioannis Pappas
- Keck School of Medicine, University of Southern California
| | | | | | | | - Robert L White
- Neurology Department, Washington University School of Medicine in St. Louis
| | - Andrew S Kayser
- Neurology Department, University of California, San Francisco
- Helen Wills Neuroscience Institute, University of California, Berkeley
- San Francisco VA Health Care System
| | | | - Mark D'Esposito
- Helen Wills Neuroscience Institute, University of California, Berkeley
- Psychology Department, University of California, Berkeley
| |
Collapse
|
2
|
Jia Y, Yang CC, Lauterborn JC, Gall CM, Wood MA, Lynch G. Cocaine Blocks Cholinergic Activity in the Medial Habenula Prior to But Not After Induced Preference for the Drug. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644247. [PMID: 40196688 PMCID: PMC11974711 DOI: 10.1101/2025.03.20.644247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Descending projections from the medial habenula potently influence brainstem systems associated with reward and mood. Relatedly, the ventral, cholinergic segment of the nucleus has been linked to nicotine and cocaine addiction. Here we report that cocaine has no effect on baseline firing in the ventral medial habenula but entirely blocks the self-sustained activity initiated by endogenous acetylcholine. This effect was not altered by antagonists to dopamine receptors and thus presumably reflects a direct action on cholinergic receptors. Remarkably, cocaine had no effect on endogenous cholinergic activity in mice that had been extinguished from an induced cocaine preference. In all, the drug has potent effects, albeit through an exotic mode of action, on the medial habenula and these are eliminated by prior experience with the drug. These results describe a novel target for cocaine that is plausibly related to the psychological effects of the drug, and an unexpected consequence of earlier use.
Collapse
|
3
|
da Silva GA, Silva LC, Filho EMT, Damasceno MV, Monte-Silva K, Ximenes-da-Silva A. Transcranial direct current stimulation associated with physical exercise can help smokers to quit smoking: a randomized controlled trial. Sci Rep 2025; 15:8623. [PMID: 40074739 PMCID: PMC11903688 DOI: 10.1038/s41598-025-85877-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
Chronic exposure to nicotine is related to low activity in the prefrontal cortex and insular hyperactivity in smokers. Therefore, addiction has been the target of experimental studies in aerobic exercise (AE) and transcranial direct current stimulation (tDCS). Thus, the objective of this study was to verify the effect of AE and anodal tDCS at F4 and cathodal at T3 on craving, motivation to change smoking behaviour (MCSB) and brain reactivity (BR) in smokers. The sample consisted of 41 chronic smokers distributed into four groups: tDCS (G1), AE (G2), tDCS combined with AE (G3) and sham tDCS combined with AE (G4). All volunteers underwent 5 consecutive sessions of the intended intervention. Before starting the intervention protocol and after the last intervention session, the volunteers answered questionnaires and underwent an electroencephalogram exam, to evaluate the variables investigated. The results demonstrated that AE, when associated with active tDCS, was effective in promoting a reduction in craving (p < 0,05), cigarette consumption (p < 0,05), and BR (p < 0,05) during exposure to smoking cues, in addition to increasing MCSB (p < 0,05). Therefore, only when associated with AE, tDCS was able to modulate positive effects on smoking.
Collapse
Affiliation(s)
- Giselma Alcantara da Silva
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (ICBS/UFAL), Maceió, Brazil
| | - Lucas Chagas Silva
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (ICBS/UFAL), Maceió, Brazil
| | - Euclides Maurício Trindade Filho
- Laboratório de Neurociências, Universidade Estadual de Ciências da Saúde de Alagoas (LABNEURO/UNCISAL), Maceió, Brazil
- Centro Universitário CESMAC, Maceió, Brazil
| | - Mayara Vieira Damasceno
- Instituto de Educação Física e Esporte, Universidade Federal de Alagoas (IEFE/UFAL), Maceió, Brazil
| | - Kátia Monte-Silva
- Laboratório de Neurociência Aplicada, Universidade Federal de Pernambuco (LANA/ UFPE, Recife, Brazil
| | - Adriana Ximenes-da-Silva
- Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas (ICBS/UFAL), Maceió, Brazil.
| |
Collapse
|
4
|
Sun J, Zhang Q, Li Y, Zhu Y, Hu N, Wang J, Mao J, Fan W, Shi Q, Chai G, Xie J. Neural modulation by nicotine aerosols and the role of flavor additives: insights from local field potentials in mice. Neuropharmacology 2025; 264:110237. [PMID: 39586494 DOI: 10.1016/j.neuropharm.2024.110237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Research on nicotine's neurobiological effects has rarely focused on aerosols, despite their primary role in tobacco product consumption. Here, we utilized in vivo electrophysiology to examine brain activity in mice exposed to nicotine aerosols, both alone and with flavor additives (citric acid and menthol). Local field potential (LFP) recordings from the nucleus accumbens (NAc), basolateral amygdala (BLA), ventral tegmental area (VTA), and ventral posteromedial nucleus (VPM) were analyzed under saline, nicotine, nicotine with citric acid(CA + NIC), and nicotine with menthol(MENT + NIC) conditions. Nicotine exposure significantly reduced power spectral density (PSD) in the NAc-Alpha, NAc-Beta, and BLA-Beta bands, unaffected by flavor additives. Coherence between key brain regions (e.g., VPM-VTA in Beta, VPM-BLA in Alpha) also decreased with nicotine but was restored with citric acid or menthol, suggesting their role in mitigating nicotine's disruptive effects on neural synchronization. Our findings show that LFPs can effectively capture nicotine's neural effects and highlight the modulatory role of flavor additives, offering new insights into nicotine exposure management and tobacco product design.
Collapse
Affiliation(s)
- Jingping Sun
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100083, PR China; Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China; Beijing Life Science Academy, Beijing, 102209, PR China
| | - Qidong Zhang
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Ying Li
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Yunhe Zhu
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Nengwei Hu
- Department of Pharmacology & Therapeutics, School of Medicine, and Institute of Neuroscience, Trinity College, Dublin 2, Ireland; Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, PR China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou, 450001, PR China
| | - Jian Mao
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Wu Fan
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Qingzhao Shi
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China
| | - Guobi Chai
- Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, Henan, 450001, PR China; Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Zhengzhou, Henan, 450001, PR China.
| | - Jianping Xie
- School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, 100083, PR China; Beijing Life Science Academy, Beijing, 102209, PR China.
| |
Collapse
|
5
|
Boumhaouad S, Makowicz EA, Choi S, Bouhaddou N, Balla J, Taghzouti K, Sulzer D, Mosharov EV. Regulation of Dopamine Release by Tonic Activity Patterns in the Striatal Brain Slice. ACS Chem Neurosci 2025; 16:303-310. [PMID: 39798080 PMCID: PMC11804867 DOI: 10.1021/acschemneuro.4c00323] [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: 05/24/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Voluntary movement, motivation, and reinforcement learning depend on the activity of ventral midbrain neurons, which extend axons to release dopamine (DA) in the striatum. These neurons exhibit two patterns of action potential activity: low-frequency tonic activity that is intrinsically generated and superimposed high-frequency phasic bursts that are driven by synaptic inputs. Ex vivo acute striatal brain preparations are widely employed to study the regulation of evoked DA release but exhibit very different DA release kinetics than in vivo recordings. To investigate the relationship between phasic and tonic neuronal activity, we stimulated the slice in patterns intended to mimic tonic activity, which were interrupted by a series of burst stimuli. Conditioning the striatal slice with low-frequency activity altered DA release triggered by high-frequency bursts and produced kinetic parameters that resemble those in vivo. In the absence of applied tonic activity, nicotinic acetylcholine receptor and D2 DA receptor antagonists had no significant effect on neurotransmitter release, driven by repeated burst activity in the striatal brain slice. In contrast, in tonically stimulated slices, the D2 receptor blockade decreased the amount of DA released during a single-burst and facilitated DA release in subsequent bursts. This experimental system provides a means to reconcile the difference in the kinetics of DA release ex vivo and in vivo and provides a novel approach to more accurately emulate pre- and postsynaptic mechanisms that control axonal DA release in vivo.
Collapse
Affiliation(s)
- Siham Boumhaouad
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Emily A Makowicz
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Sejoon Choi
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Nezha Bouhaddou
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Jihane Balla
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Khalid Taghzouti
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - David Sulzer
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Eugene V. Mosharov
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| |
Collapse
|
6
|
Malik R, Beaton D, Ahmed J, Nho K, Saykin AJ, Wang J, Hegele RA, Finger E. A DAT1 gene and APOE ε4 interaction is associated with apathy and structural brain changes in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis 2025; 103:230-242. [PMID: 39610284 PMCID: PMC12028270 DOI: 10.1177/13872877241299785] [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] [Indexed: 11/30/2024]
Abstract
BACKGROUND Apathy in patients with Alzheimer's disease (AD) is associated with significant morbidity and is often one of the first neuropsychiatric symptoms to present in mild cognitive impairment (MCI). Apathy is associated with accelerated cognitive decline and atrophy in fronto-striatal regions of the brain. Previous work has shown a link between apathy and the APOE gene in the context of AD, as the APOE ε4 allele is already known to be associated with the onset of AD. However, other genetic associations with apathy are largely unexplored. OBJECTIVE To examine whether interactions between genetic variants related to neurotransmitter systems and regional brain atrophy are associated with apathy in patients with MCI and AD. METHODS In a sample of individuals with AD (n = 266), MCI (n = 518), and cognitively normal controls (n = 378), a partial least squares correspondence analysis modeled interactions between single nucleotide polymorphisms, structural whole-brain imaging variables, and apathy. RESULTS An interaction was found between apathy, the possession of an APOE ε4 allele combined with minor homozygosity for the DAT1 (dopamine transporter 1) gene, and regional brain atrophy. This interaction was closely linked to the MCI and AD groups. CONCLUSIONS The results point to an association of a dopaminergic genetic marker and apathy in the AD continuum and may inform future design of clinical trials of apathy, as well as new treatment targets.
Collapse
Affiliation(s)
- Rubina Malik
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Canada
| | - Derek Beaton
- Data Science & Advanced Analytics, Unity Health Toronto, Toronto, ON, Canada
| | - Juweiriya Ahmed
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jian Wang
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Robert A Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Canada
| |
Collapse
|
7
|
Francis AN, Sebille S, Whitfield-Gabrieli S, Camprodon JA. Multimodal 7T imaging reveals enhanced functional coupling between salience and frontoparietal networks in young adult tobacco cigarette smokers. Brain Imaging Behav 2024; 18:913-921. [PMID: 38639847 DOI: 10.1007/s11682-024-00882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/20/2024]
Abstract
Tobacco cigarette smoking is associated with disrupted brain network dynamics in resting brain networks including the Salience (SN) and Fronto parietal (FPN). Unified multimodal methods [Resting state connectivity analysis, Diffusion Tensor Imaging (DTI), neurite orientation dispersion and density imaging (NODDI), and cortical thickness analysis] were employed to test the hypothesis that the impact of cigarette smoking on the balance among these networks is due to alterations in white matter connectivity, microstructural architecture, functional connectivity and cortical thickness (CT) and that these metrics define fundamental differences between people who smoke and nonsmokers. Multimodal analyses of previously collected 7 Tesla MRI data via the Human Connectome Project were performed on 22 people who smoke (average number of daily cigarettes was 10 ± 5) and 22 age- and sex-matched nonsmoking controls. First, functional connectivity analysis was used to examine SN-FPN-DMN interactions between people who smoke and nonsmokers. The anatomy of these networks was then assessed using DTI and CT analyses while microstructural architecture of WM was analyzed using the NODDI toolbox. Seed-based connectivity analysis revealed significantly enhanced within network [p = 0.001 FDR corrected] and between network functional coupling of the salience and R-frontoparietal networks in people who smoke [p = 0.004 FDR corrected]. The network connectivity was lateralized to the right hemisphere. Whole brain diffusion analysis revealed no significant differences between people who smoke and nonsmokers in Fractional Anisotropy, Mean diffusivity and in neurite orienting and density. There were also no significant differences in CT in the hubs of these networks. Our results demonstrate that tobacco cigarette smoking is associated with enhanced functional connectivity, but anatomy is largely intact in young adults. Whether this enhanced connectivity is pre-existing, transient or permanent is not known. The observed enhanced connectivity in resting state networks may contribute to the maintenance of smoking frequency.
Collapse
Affiliation(s)
- Alan N Francis
- Department of Neuroscience, University of Texas, Rio Grande Valley, Edinburg, TX, USA.
| | - Sophie Sebille
- Department of Neuroscience GHU Paris Psychiatrie et Neurosciences, Paris, France
| | | | - Joan A Camprodon
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
8
|
Williams BM, Steed ND, Woolley JT, Moedl AA, Nelson CA, Jones GC, Burris MD, Arias HR, Kim OH, Jang EY, Hone AJ, McIntosh JM, Yorgason JT, Steffensen SC. Catharanthine Modulates Mesolimbic Dopamine Transmission and Nicotine Psychomotor Effects via Inhibition of α6-Nicotinic Receptors and Dopamine Transporters. ACS Chem Neurosci 2024; 15:1738-1754. [PMID: 38613458 PMCID: PMC11744774 DOI: 10.1021/acschemneuro.3c00478] [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] [Indexed: 04/15/2024] Open
Abstract
Iboga alkaloids, also known as coronaridine congeners, have shown promise in the treatment of alcohol and opioid use disorders. The objective of this study was to evaluate the effects of catharanthine and 18-methoxycoronaridine (18-MC) on dopamine (DA) transmission and cholinergic interneurons in the mesolimbic DA system, nicotine-induced locomotor activity, and nicotine-taking behavior. Utilizing ex vivo fast-scan cyclic voltammetry (FSCV) in the nucleus accumbens core of male mice, we found that catharanthine or 18-MC differentially inhibited evoked DA release. Catharanthine inhibition of evoked DA release was significantly reduced by both α4 and α6 nicotinic acetylcholine receptors (nAChRs) antagonists. Additionally, catharanthine substantially increased DA release more than vehicle during high-frequency stimulation, although less potently than an α4 nAChR antagonist, which confirms previous work with nAChR antagonists. Interestingly, while catharanthine slowed DA reuptake measured via FSCV ex vivo, it also increased extracellular DA in striatal dialysate from anesthetized mice in vivo in a dose-dependent manner. Superfusion of catharanthine or 18-MC inhibited the firing rate of striatal cholinergic interneurons in a concentration dependent manner, which are known to potently modulate presynaptic DA release. Catharanthine or 18-MC suppressed acetylcholine currents in oocytes expressing recombinant rat α6/α3β2β3 or α6/α3β4 nAChRs. In behavioral experiments using male Sprague-Dawley rats, systemic administration of catharanthine or 18-MC blocked nicotine enhancement of locomotor activity. Importantly, catharanthine attenuated nicotine self-administration in a dose-dependent manner while having no effect on food reinforcement. Lastly, administration of catharanthine and nicotine together greatly increased head twitch responses, indicating a potential synergistic hallucinogenic effect. These findings demonstrate that catharanthine and 18-MC have similar, but not identical effects on striatal DA dynamics, striatal cholinergic interneuron activity and nicotine psychomotor effects.
Collapse
Affiliation(s)
- Benjamin M. Williams
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Nathan D. Steed
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Joel T. Woolley
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Aubrey A. Moedl
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Christina A. Nelson
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Gavin C. Jones
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Matthew D. Burris
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Hugo R. Arias
- Department of Pharmacology and Physiology, Oklahoma State University College of Osteopathic Medicine, Tahlequah, Oklahoma 74464, United States
| | - Oc-Hee Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Eun Young Jang
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Arik J. Hone
- George E. Wahlen Veterans Affairs Medical Center, and Departments of Psychiatry and Biology, University of Utah, Salt Lake City, Utah 84112, United States
| | - J. Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, and Departments of Psychiatry and Biology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jordan T. Yorgason
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| | - Scott C. Steffensen
- Department of Psychology/Neuroscience, Brigham Young University, Provo, Utah 84602, United States
| |
Collapse
|
9
|
Miller G, Pareek O, Penman SL, Thanos PK. The Effects of Nicotine and Cannabinoids on Cytokines. Curr Pharm Des 2024; 30:2468-2484. [PMID: 38859790 DOI: 10.2174/0113816128293077240529111824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND The usage of nicotine and cannabinoids has rapidly grown in popularity, leading to increased research into how they can affect people's health, both positively and negatively. Nicotine, Cannabidiol (CBD), and Δ9-tetrahydrocannabinol (THC) have been shown to have significant effects on cytokine function and inflammatory response. OBJECTIVE This study aimed to review and summarize the current literature on the effects of nicotine and cannabinoids on cytokines, including interleukins, TNF, IFN, and TGF-β. METHODS Literature search was conducted on Medline/PubMed electronic databases utilizing the search terms "nicotine" OR "cannabis" OR "cannabinoids" AND "cytokine" AND "inflammation" AND "stress" AND "immune" from 11/1973 to 02/2024. RESULTS THC and CBD usage have been associated with conflicting impacts on immune response, and observed to both exacerbate and inhibit inflammation. Nicotine has been shown to be generally proinflammatory with regards to cytokines. These responses have been reported to have significant effects on bodily response to inflammation-related diseases. Nicotine usage is associated with worsened outcomes for some conditions, like chronic pain, but improved outcomes for others, like arthritis. The impacts of cannabinoid usage tend to be more positive, exerting anti-inflammatory effects across a wide range of diseases. Given the widespread usage of these substances, it is important to understand the nature of their consequences on immune functions and the underlying mechanisms by which they act. CONCLUSION This review has covered how cannabinoids and nicotine affect inflammation directly and how these effects can be attributed to the treatment of inflammatory diseases. In summary, the existing research studying the effects of cannabinoids and nicotine supports the major relationship between nicotine and cannabis use and inflammatory diseases.
Collapse
Affiliation(s)
- Grace Miller
- Department of Pharmacology Toxicology, University at Buffalo, State University of New York, Buffalo, NY 14068, United States
| | - Ojas Pareek
- Department of Pharmacology Toxicology, University at Buffalo, State University of New York, Buffalo, NY 14068, United States
| | - Samantha L Penman
- Department of Pharmacology Toxicology, University at Buffalo, State University of New York, Buffalo, NY 14068, United States
| | - Panayotis K Thanos
- Department of Pharmacology Toxicology, University at Buffalo, State University of New York, Buffalo, NY 14068, United States
| |
Collapse
|
10
|
Luo Y, Yang Y, Schneider C, Balle T. The Anti-Nociceptive Effects of Nicotine in Humans: A Systematic Review and Meta-Analysis. Pharmaceuticals (Basel) 2023; 16:1665. [PMID: 38139792 PMCID: PMC10747127 DOI: 10.3390/ph16121665] [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: 08/18/2023] [Revised: 10/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Pain can have a serious impact on a patient's physical, mental, and social health, often causing their quality of life to decline. Various nicotine dosage forms, such as nicotine patches and nasal spray, have been developed and used as analgesics in clinical settings. However, there is controversy over the anti-nociceptive effects of nicotine among different clinical trials. The purpose of this meta-analysis is to quantify the analgesic effect of nicotine patches, nicotine nasal spray, and tobacco smoking on pain in humans. METHODS Relevant articles published in English prior to July 2023 were identified using the PubMed, Cochrane Library, and Embase online databases in accordance with PRISMA (2020) guidelines. Two reviewers independently screened and selected studies, extracted data, and assessed the quality of the included studies using version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB 2). RStudio was used for data synthesis, heterogeneity assessment, sensitivity analysis, publication bias assessment, trim-and-fill analyses, and generating forest plots. RESULTS Sixteen eligible articles, including k = 5 studies of pain tolerance (n = 210), k = 5 studies of pain threshold (n = 210), and k = 12 studies of pain scores (N = 1249), were included for meta-analysis. Meta-analytic integration for pain threshold (Hedges' g = 0.28, 95% CI = 0-0.55, Z = 1.99, p = 0.05) and pain tolerance (Hedges' g = 0.32, 95% CI = 0.05-0.59, Z = 2.30, p = 0.02) revealed that nicotine administered via tobacco smoke generated acute analgesic effects to thermal stimuli. Meta-analytic integration for pain scores revealed that nicotine had a weak anti-nociceptive effect on postoperative pain of -0.37 (95% CI = -0.77 to 0.03, Z = -1.80) but with no statistical significance (p = 0.07). In addition, a limited number of included studies revealed that long-term smoking produced hyperalgesia that may be characterized as small to medium in magnitude (Hedges' g = 0.37, 95% CI = 0.29-0.64, Z = 5.33, p < 0.01). CONCLUSION These results help to clarify the mixed outcomes of trials and may ultimately inform the treatment of pain. We observed that acute nicotine administration prolonged the laboratory-induced pain threshold and tolerance time and may mildly relieve postoperative pain. In addition, long-term tobacco smoking may have a nociceptive effect on different types of chronic pain. More research is needed to determine the anti-nociceptive effects of nicotine in humans, and to understand the optimal timing, dose, and method of delivery of nicotine.
Collapse
Affiliation(s)
- Yujia Luo
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (Y.L.)
- Brain and Mind Centre, The University of Sydney, 94 Mallet Street, Camperdown, NSW 2050, Australia
| | - Yating Yang
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (Y.L.)
- Brain and Mind Centre, The University of Sydney, 94 Mallet Street, Camperdown, NSW 2050, Australia
| | - Carl Schneider
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (Y.L.)
| | - Thomas Balle
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (Y.L.)
- Brain and Mind Centre, The University of Sydney, 94 Mallet Street, Camperdown, NSW 2050, Australia
| |
Collapse
|
11
|
Han P, Jing X, Han S, Wang X, Li Q, Zhang Y, Yu P, Liu XA, Wu P, Chen H, Hou H, Hu Q. Pharmacokinetic differences in nicotine and nicotine salts mediate reinforcement-related behavior: an animal model study. Front Neurosci 2023; 17:1288102. [PMID: 38033549 PMCID: PMC10687399 DOI: 10.3389/fnins.2023.1288102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Since their introduction in the United States and Europe in 2007, electronic cigarettes (E-Cigs) have become increasingly popular among smokers. Nicotine, a key component in both tobacco and e-cigarettes, can exist in two forms: nicotine-freebase (FBN) and nicotine salts (NS). While nicotine salt is becoming more popular in e-cigarettes, the effect of nicotine salts on reinforcement-related behaviors remains poorly understood. This study aimed to compare the reinforcing effects of nicotine and nicotine salts in animal models of drug self-administration and explore potential mechanisms that may contribute to these differences. The results demonstrated that three nicotine salts (nicotine benzoate, nicotine lactate, and nicotine tartrate) resulted in greater reinforcement-related behaviors in rats compared to nicotine-freebase. Moreover, withdrawal-induced anxiety symptoms were lower in the three nicotine salt groups than in the nicotine-freebase group. The study suggested that differences in the pharmacokinetics of nicotine-freebase and nicotine salts in vivo may explain the observed behavioral differences. Overall, this study provides valuable insights into the reinforcing effects of nicotine as well as potential differences between nicotine-freebase and nicotine salts.
Collapse
Affiliation(s)
- Pengfei Han
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, China
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xiaoyuan Jing
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shulei Han
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xinsheng Wang
- Institute of Biomedical Engineering, College of Life Sciences, Qingdao University, Qingdao, China
| | - Qiannan Li
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Yuan Zhang
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Pengpeng Yu
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xin-an Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ping Wu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Beijing, China
| | - Huan Chen
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Qingyuan Hu
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| |
Collapse
|
12
|
Wadsworth HA, Anderson EQ, Williams BM, Ronström JW, Moen JK, Lee AM, McIntosh JM, Wu J, Yorgason JT, Steffensen SC. Role of α6-Nicotinic Receptors in Alcohol-Induced GABAergic Synaptic Transmission and Plasticity to Cholinergic Interneurons in the Nucleus Accumbens. Mol Neurobiol 2023; 60:3113-3129. [PMID: 36802012 PMCID: PMC10690621 DOI: 10.1007/s12035-023-03263-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/03/2023] [Indexed: 02/21/2023]
Abstract
The prevailing view is that enhancement of dopamine (DA) transmission in the mesolimbic system, consisting of DA neurons in the ventral tegmental area (VTA) that project to the nucleus accumbens (NAc), underlies the reward properties of ethanol (EtOH) and nicotine (NIC). We have shown previously that EtOH and NIC modulation of DA release in the NAc is mediated by α6-containing nicotinic acetylcholine receptors (α6*-nAChRs), that α6*-nAChRs mediate low-dose EtOH effects on VTA GABA neurons and EtOH preference, and that α6*-nAChRs may be a molecular target for low-dose EtOH. However, the most sensitive target for reward-relevant EtOH modulation of mesolimbic DA transmission and the involvement of α6*-nAChRs in the mesolimbic DA reward system remains to be elucidated. The aim of this study was to evaluate EtOH effects on GABAergic modulation of VTA GABA neurons and VTA GABAergic input to cholinergic interneurons (CINs) in the NAc. Low-dose EtOH enhanced GABAergic input to VTA GABA neurons that was blocked by knockdown of α6*-nAChRs. Knockdown was achieved either by α6-miRNA injected into the VTA of VGAT-Cre/GAD67-GFP mice or by superfusion of the α-conotoxin MII[H9A;L15A] (MII). Superfusion of MII blocked EtOH inhibition of mIPSCs in NAc CINs. Concomitantly, EtOH enhanced CIN firing rate, which was blocked by knockdown of α6*-nAChRs with α6-miRNA injected into the VTA of VGAT-Cre/GAD67-GFP mice. The firing rate of CINs was not enhanced by EtOH in EtOH-dependent mice, and low-frequency stimulation (LFS; 1 Hz, 240 pulses) caused inhibitory long-term depression at this synapse (VTA-NAc CIN-iLTD) which was blocked by knockdown of α6*-nAChR and MII. Ethanol inhibition of CIN-mediated evoked DA release in the NAc was blocked by MII. Taken together, these findings suggest that α6*-nAChRs in the VTA-NAc pathway are sensitive to low-dose EtOH and play a role in plasticity associated with chronic EtOH.
Collapse
Affiliation(s)
- Hillary A Wadsworth
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA
| | - Elizabeth Q Anderson
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA
| | - Benjamin M Williams
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA
| | - Joakim W Ronström
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA
| | - Janna K Moen
- Department of Pharmacology, Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anna M Lee
- Department of Pharmacology, Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - J Michael McIntosh
- School of Biological Sciences and Department of Psychiatry, University of Utah, Salt Lake City, UT, 84108, USA
- George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, 84148, USA
| | - Jie Wu
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jordan T Yorgason
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA
| | - Scott C Steffensen
- Department of Psychology and Neuroscience, Brigham Young University, 1050 KMBL, Provo, UT, 84602, USA.
| |
Collapse
|
13
|
Fatty Acid-Binding Protein 5 Gene Deletion Enhances Nicotine-Conditioned Place Preference: Illuminating the Putative Gateway Mechanisms. FUTURE PHARMACOLOGY 2023; 3:108-116. [PMID: 36864947 PMCID: PMC9969817 DOI: 10.3390/futurepharmacol3010007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Emerging evidence indicates that the endogenous cannabinoid system modulates the behavioral and physiological effects of nicotine. Fatty acid-binding proteins (FABPs) are among the primary intracellular trafficking mechanisms of endogenous cannabinoids, such as anandamide. To this end, changes in FABP expression may similarly impact the behavioral manifestations associated with nicotine, particularly its addictive properties. FABP5 +/+ and FABP5 -/- mice were tested for nicotine-conditioned place preference (CPP) at two different doses (0.1 or 0.5 mg/kg). The nicotine-paired chamber was assigned as their least preferred chamber during preconditioning. Following 8 days of conditioning, the mice were injected with either nicotine or saline. The mice were allowed to access to all the chambers on the test day, and their times spent in the drug chamber on the preconditioning versus the test days were used to examine the drug preference score. The CPP results showed that the FABP5 -/- mice displayed a higher place preference for 0.1 mg/kg nicotine than the FABP5 +/+ mice, while no CPP difference was observed for 0.5 mg/kg nicotine between the genotypes. In conclusion, FABP5 plays an important role in regulating nicotine place preference. Further research is warranted to identify the precise mechanisms. The results suggest that dysregulated cannabinoid signaling may impact nicotine-seeking behavior.
Collapse
|
14
|
Neurobiology and Mechanisms of Nicotine Addiction. Respir Med 2023. [DOI: 10.1007/978-3-031-24914-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
15
|
Stowe TA, Pitts EG, Leach AC, Iacino MC, Niere F, Graul B, Raab-Graham KF, Yorgason JT, Ferris MJ. Diurnal rhythms in cholinergic modulation of rapid dopamine signals and associative learning in the striatum. Cell Rep 2022; 39:110633. [PMID: 35385720 PMCID: PMC9148619 DOI: 10.1016/j.celrep.2022.110633] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/20/2021] [Accepted: 03/16/2022] [Indexed: 11/26/2022] Open
Abstract
Dysregulation of biological rhythms plays a role in a wide range of psychiatric disorders. We report mechanistic insights into the rhythms of rapid dopamine signals and cholinergic interneurons (CINs) working in concert in the rodent striatum. These rhythms mediate diurnal variation in conditioned responses to reward-associated cues. We report that the dopamine signal-to-noise ratio varies according to the time of day and that phasic signals are magnified during the middle of the dark cycle in rats. We show that CINs provide the mechanism for diurnal variation in rapid dopamine signals by serving as a gain of function to the dopamine signal-to-noise ratio that adjusts across time of day. We also show that conditioned responses to reward-associated cues exhibit diurnal rhythms, with cue-directed behaviors observed exclusively midway through the dark cycle. We conclude that the rapid dopamine signaling rhythm is mediated by a diurnal rhythm in CIN activity, which influences learning and motivated behaviors across the time of day.
Collapse
Affiliation(s)
- Taylor A Stowe
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Elizabeth G Pitts
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Amy C Leach
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Melody C Iacino
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Farr Niere
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Benjamin Graul
- Department of Cellular Biology and Physiology, Neuroscience Center, Brigham Young University, Provo, UT 84602, USA
| | - Kimberly F Raab-Graham
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Jordan T Yorgason
- Department of Cellular Biology and Physiology, Neuroscience Center, Brigham Young University, Provo, UT 84602, USA
| | - Mark J Ferris
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA.
| |
Collapse
|
16
|
Leach AC, Pitts EG, Siciliano CA, Ferris MJ. α7 nicotinic acetylcholine receptor modulation of accumbal dopamine release covaries with novelty seeking. Eur J Neurosci 2022; 55:1162-1173. [PMID: 35141983 PMCID: PMC9586210 DOI: 10.1111/ejn.15620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
Heightened novelty-seeking phenotypes are associated with a range of behavioural traits including susceptibility to drug use. These relationships are recapitulated in preclinical models, where rats that exhibit increased exploratory activity in novel environments (high responders-HR) acquire self-administration of psychostimulants more rapidly compared to rats that display low novelty exploration (low responders-LR). Dopamine release dynamics in the nucleus accumbens (NAc) covaries with response to novelty, and differences in dopaminergic signalling are thought to be a major underlying driver of the link between novelty seeking and drug use vulnerability. Accumbal dopamine release is controlled by local microcircuits including modulation through glutamatergic and nicotinic acetylcholine receptor (nAChR) systems, but whether these mechanisms contribute to disparate dopamine signalling across novelty phenotypes is unclear. Here, we used ex vivo voltammetry in the NAc of rats to determine if α7 nAChRs contribute to differential dopamine dynamics associated with individual differences in novelty exploration. We found that blockade of α7 nAChRs attenuates tonic dopamine release evoked by low-frequency stimulations across phenotypes but that phasic release is decreased in LRs while HRs are unaffected. These stimulation frequency- and phenotype-dependent effects result in a decreased dynamic range of release exclusively in LRs. Furthermore, we found that differential α7 modulation of dopamine release in LRs is dependent on AMPA but not NMDA receptors. These results help to form an understanding of the local NAc microcircuitry and provide a potential mechanism for covariance of dopamine dynamics and sensitivity to the reinforcing effects of drugs of abuse.
Collapse
Affiliation(s)
- Amy C. Leach
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Elizabeth G. Pitts
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Cody A. Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN
| | - Mark J. Ferris
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
17
|
Downs AM, Donsante Y, Jinnah H, Hess EJ. Blockade of M4 muscarinic receptors on striatal cholinergic interneurons normalizes striatal dopamine release in a mouse model of TOR1A dystonia. Neurobiol Dis 2022; 168:105699. [DOI: 10.1016/j.nbd.2022.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/10/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022] Open
|
18
|
Goldstein N, Carty JRE, Betley JN. Specificity of Varenicline in Blocking Mesolimbic Circuit Activation to Natural and Drug Rewards. Neuroscience 2022; 483:40-51. [PMID: 34923039 PMCID: PMC8837713 DOI: 10.1016/j.neuroscience.2021.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022]
Abstract
The mesolimbic dopamine (DA) system reinforces behaviors that are critical for survival. However, drug dependence can occur when drugs of abuse, such as nicotine, highjack this reinforcement system. Pharmacologically targeting the DA system to selectively block drug reinforcement requires a detailed understanding of the neural circuits and molecular pathways that lead to the reward-based activation of mesolimbic circuits. Varenicline is an approved smoking cessation drug that has been shown to block nicotine-evoked DA increases in the nucleus accumbens (NAc) through action on nicotinic acetylcholine receptors. Because these receptors have been implicated in the reinforcement of other addictive substances, we explored the possibility that varenicline could broadly affect reward processing. We used in vivo fiber photometry to monitor midbrain DA neuron activity and striatal DA levels following either natural or drug rewards in mice treated with varenicline. We demonstrate that varenicline pretreatment enhances the suppression of nicotine-evoked DA release by attenuating DA neuron activity in the VTA. Varenicline's ability to attenuate DA release is highly specific to nicotine, and varenicline slightly elevates DA release when co-administered with morphine or ethanol. Furthermore, varenicline has no effect on DA release in response to naturally rewarding behavior such as food intake or exercise. These results demonstrate the exquisite specificity with which varenicline blocks nicotine reward and highlight the complexity with which different rewards activate the mesolimbic DA system.
Collapse
Affiliation(s)
- Nitsan Goldstein
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jamie R E Carty
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
19
|
Lin L, Lan Y, Zhu H, Yu L, Wu S, Wan W, Shu Y, Xiang H, Hou T, Zhang H, Ma Y, Su W, Li M. Effects of Chemogenetic Inhibition of D1 or D2 Receptor-Containing Neurons of the Substantia Nigra and Striatum in Mice With Tourette Syndrome. Front Mol Neurosci 2021; 14:779436. [PMID: 34955745 PMCID: PMC8696039 DOI: 10.3389/fnmol.2021.779436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
As tourette syndrome (TS) is a common neurobehavioral disorder, the primary symptoms of which include behavioral stereotypies. Dysfunction of the substantia nigra-striatum network could be the main pathogenesis of TS, which is closely associated with dopamine (DA) and its receptors. TS is often resistant to conventional treatments. Therefore, it is necessary to investigate the neurobiological mechanisms underlying its pathogenesis. In this study, we investigated whether chemogenetic activation or inhibition of dopaminergic D1 receptor (D1R)- or D2 receptor (D2R)-containing neurons in the substantia nigra pars compacta (SNpc) or dorsal striatum (dSTR) affected the stereotyped behavior and motor functions of TS mice. Intraperitoneal injection of 3,3'-iminodipropionitrile (IDPN) was used to induce TS in mice. Stereotyped behavior test and open-field, rotarod, and grip strength tests were performed to evaluate stereotyped behavior and motor functions, respectively. Immunofluorescence labeling was used to detect the co-labeling of virus fluorescence and D1R or D2R. We found that chemogenetic inhibition of D1R- or D2R-containing neurons in the SNpc and dSTR alleviated behavioral stereotypies and motor functions in TS mice. Chemogenetic activation of D1R-containing neurons in the dSTR aggravated behavioral stereotypies and motor functions in vehicle-treated mice, but neither was aggravated in TS mice. In conclusion, chemogenetic inhibition of D1R- or D2R-containing neurons in the SNpc and dSTR alleviated behavioral stereotypies of TS, providing a new treatment target for TS. Moreover, the activation of D1R-containing neurons in the dSTR may contribute to the pathogenesis of TS, which can be chosen as a more precise target for treatment.
Collapse
Affiliation(s)
- Lixue Lin
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Rehabilitation, Wuhan No.1 Hospital, Wuhan, China
| | - Yuye Lan
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - He Zhu
- Institute of Clinical Medicine, Zhanjiang Central People's Hospital, Zhanjiang, China
| | - Lingling Yu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wangyixuan Wan
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Shu
- Department of Central Laboratory, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hongchun Xiang
- Department of Acupuncture and Moxibustion, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tengfei Hou
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ma
- Department of Rehabilitation, Wuhan No.1 Hospital, Wuhan, China
| | - Wen Su
- Department of Pediatrics, Wuhan No.1 Hospital, Wuhan, China
| | - Man Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Ciccocioppo R, de Guglielmo G, Li HW, Melis M, Caffino L, Shen Q, Domi A, Fumagalli F, Demopulos GA, Gaitanaris GA. Selective Inhibition of Phosphodiesterase 7 Enzymes Reduces Motivation for Nicotine Use through Modulation of Mesolimbic Dopaminergic Transmission. J Neurosci 2021; 41:6128-6143. [PMID: 34083258 PMCID: PMC8276738 DOI: 10.1523/jneurosci.3180-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Approximately 5 million people die from diseases related to nicotine addiction and tobacco use each year. The nicotine-induced increase of corticomesolimbic dopaminergic (DAergic) transmission and hypodopaminergic conditions occurring during abstinence are important for maintaining drug-use habits. We examined the notion of reequilibrating DAergic transmission by inhibiting phosphodiesterase 7 (PDE7), an intracellular enzyme highly expressed in the corticomesolimbic circuitry and responsible for the degradation of cyclic adenosine monophosphate (cAMP), the main second messenger modulated by DA receptor activation. Using selective PDE7 inhibitors, we demonstrated in male rats that systemic PDE7 enzyme inhibition reduced nicotine self-administration and prevented reinstatement to nicotine seeking evoked by cues or by the pharmacological stressor yohimbine. The effect was also observed by direct application of the PDE7 inhibitors into the nucleus accumbens (NAc) shell but not into the core. Inhibition of PDE7 resulted in increased DA- and cAMP-regulated neuronal phosphoprotein and cAMP response element-binding protein and their phosphorylated forms in the NAc. It also enhanced the DA D1 receptor agonism-mediated effects, indicating potentiation of protein kinase A-dependent transmission downstream of D1 receptor activation. In electrophysiological recordings from DA neurons in the lateral posterior ventral tegmental area, the PDE7 inhibitors attenuated the spontaneous activity of DA neurons. This effect was exerted through the potentiation of D1 receptor signaling and the subsequent facilitation of γ-aminobutyric acid transmission. The PDE7 inhibitors did not elicit conditioned place preference and did not induce intravenous self-administration, indicating lack of reinforcing properties. Thus, PDE7 inhibitors have the potential to treat nicotine abuse.SIGNIFICANCE STATEMENT The World Health Organization estimates that there are 1.25 billion smokers worldwide, representing one-third of the global population over the age of 15. Nicotine-induced increase of corticomesolimbic DAergic transmission and hypodopaminergic conditions occurring during abstinence are critical for maintaining drug-use habits. Here, we demonstrate that nicotine consumption and relapse to nicotine seeking are attenuated by reequilibrating DAergic transmission through inhibition of PDE7, an intracellular enzyme responsible for the degradation of cAMP, the main second messenger modulated by DA receptor activation. PDE7 inhibition may represent a novel treatment approach to aid smoking cessation.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Hong Wu Li
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Quienwei Shen
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Ana Domi
- School of Pharmacy, Pharmacology Unit, University of Camerino, 62032 Camerino, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | | | | |
Collapse
|
21
|
Wills L, Kenny PJ. Addiction-related neuroadaptations following chronic nicotine exposure. J Neurochem 2021; 157:1652-1673. [PMID: 33742685 DOI: 10.1111/jnc.15356] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022]
Abstract
The addiction-relevant molecular, cellular, and behavioral actions of nicotine are derived from its stimulatory effects on neuronal nicotinic acetylcholine receptors (nAChRs) in the central nervous system. nAChRs expressed by dopamine-containing neurons in the ventral midbrain, most notably in the ventral tegmental area (VTA), contribute to the reward-enhancing properties of nicotine that motivate the use of tobacco products. nAChRs are also expressed by neurons in brain circuits that regulate aversion. In particular, nAChRs expressed by neurons in the medial habenula (mHb) and the interpeduncular nucleus (IPn) to which the mHb almost exclusively projects regulate the "set-point" for nicotine aversion and control nicotine intake. Different nAChR subtypes are expressed in brain reward and aversion circuits and nicotine intake is titrated to maximally engage reward-enhancing nAChRs while minimizing the recruitment of aversion-promoting nAChRs. With repeated exposure to nicotine, reward- and aversion-related nAChRs and the brain circuits in which they are expressed undergo adaptations that influence whether tobacco use will transition from occasional to habitual. Genetic variation that influences the sensitivity of addiction-relevant brain circuits to the actions of nicotine also influence the propensity to develop habitual tobacco use. Here, we review some of the key advances in our understanding of the mechanisms by which nicotine acts on brain reward and aversion circuits and the adaptations that occur in these circuits that may drive addiction to nicotine-containing tobacco products.
Collapse
Affiliation(s)
- Lauren Wills
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| |
Collapse
|
22
|
Han Y, Xia G, He Y, He Y, Farias M, Xu Y, Wu Q. A hindbrain dopaminergic neural circuit prevents weight gain by reinforcing food satiation. SCIENCE ADVANCES 2021; 7:eabf8719. [PMID: 34039606 DOI: 10.1126/sciadv.abf8719] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
The neural circuitry mechanism that underlies dopaminergic (DA) control of innate feeding behavior is largely uncharacterized. Here, we identified a subpopulation of DA neurons situated in the caudal ventral tegmental area (cVTA) directly innervating DRD1-expressing neurons within the lateral parabrachial nucleus (LPBN). This neural circuit potently suppresses food intake via enhanced satiation response. Notably, this cohort of DAcVTA neurons is activated immediately before the cessation of each feeding bout. Acute inhibition of these DA neurons before bout termination substantially suppresses satiety and prolongs the consummatory feeding. Activation of postsynaptic DRD1LPBN neurons inhibits feeding, whereas genetic deletion of Drd1 within the LPBN causes robust increase in food intake and subsequent weight gain. Furthermore, the DRD1LPBN signaling manifests the central mechanism in methylphenidate-induced hypophagia. In conclusion, our study illuminates a hindbrain DAergic circuit that controls feeding through dynamic regulation in satiety response and meal structure.
Collapse
Affiliation(s)
- Yong Han
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Guobin Xia
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Monica Farias
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Qi Wu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
23
|
Wittenberg RE, Wolfman SL, De Biasi M, Dani JA. Nicotinic acetylcholine receptors and nicotine addiction: A brief introduction. Neuropharmacology 2020; 177:108256. [PMID: 32738308 PMCID: PMC7554201 DOI: 10.1016/j.neuropharm.2020.108256] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 12/13/2022]
Abstract
Nicotine is a highly addictive drug found in tobacco that drives its continued use despite the harmful consequences. The initiation of nicotine abuse involves the mesolimbic dopamine system, which contributes to the rewarding sensory stimuli and associative learning processes in the beginning stages of addiction. Nicotine binds to neuronal nicotinic acetylcholine receptors (nAChRs), which come in a diverse collection of subtypes. The nAChRs that contain the α4 and β2 subunits, often in combination with the α6 subunit, are particularly important for nicotine's ability to increase midbrain dopamine neuron firing rates and phasic burst firing. Chronic nicotine exposure results in numerous neuroadaptations, including the upregulation of particular nAChR subtypes associated with long-term desensitization of the receptors. When nicotine is no longer present, for example during attempts to quit smoking, a withdrawal syndrome develops. The expression of physical withdrawal symptoms depends mainly on the α2, α3, α5, and β4 nicotinic subunits in the epithalamic habenular complex and its target regions. Thus, nicotine affects diverse neural systems and an array of nAChR subtypes to mediate the overall addiction process. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Ruthie E Wittenberg
- Departments of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shannon L Wolfman
- Departments of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariella De Biasi
- Departments of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Psychiatry, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John A Dani
- Departments of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Caton M, Ochoa ELM, Barrantes FJ. The role of nicotinic cholinergic neurotransmission in delusional thinking. NPJ SCHIZOPHRENIA 2020; 6:16. [PMID: 32532978 PMCID: PMC7293341 DOI: 10.1038/s41537-020-0105-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
Delusions are a difficult-to-treat and intellectually fascinating aspect of many psychiatric illnesses. Although scientific progress on this complex topic has been challenging, some recent advances focus on dysfunction in neural circuits, specifically in those involving dopaminergic and glutamatergic neurotransmission. Here we review the role of cholinergic neurotransmission in delusions, with a focus on nicotinic receptors, which are known to play a part in some illnesses where these symptoms appear, including delirium, schizophrenia spectrum disorders, bipolar disorder, Parkinson, Huntington, and Alzheimer diseases. Beginning with what we know about the emergence of delusions in these illnesses, we advance a hypothesis of cholinergic disturbance in the dorsal striatum where nicotinic receptors are operative. Striosomes are proposed to play a central role in the formation of delusions. This hypothesis is consistent with our current knowledge about the mechanism of action of cholinergic drugs and with our abstract models of basic cognitive mechanisms at the molecular and circuit levels. We conclude by pointing out the need for further research both at the clinical and translational levels.
Collapse
Affiliation(s)
- Michael Caton
- The Permanente Medical Group, Kaiser Santa Rosa Department of Psychiatry, 2235 Mercury Way, Santa Rosa, CA, 95047, USA
- Heritage Oaks Hospital, 4250 Auburn Boulevard, Sacramento, CA, 95841, USA
| | - Enrique L M Ochoa
- Heritage Oaks Hospital, 4250 Auburn Boulevard, Sacramento, CA, 95841, USA
- Volunteer Clinical Faculty, Department of Psychiatry and Behavioral Sciences, University of California at Davis, 2230 Stockton Boulevard, Sacramento, CA, 95817, USA
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute for Biomedical Research (BIOMED), Faculty of Medical Sciences, UCA-CONICET, Av. Alicia Moreau de Justo 1600, C1107AFF, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Wang KS, Zegel M, Molokotos E, Moran LV, Olson DP, Pizzagalli DA, Janes AC. The acute effects of nicotine on corticostriatal responses to distinct phases of reward processing. Neuropsychopharmacology 2020; 45:1207-1214. [PMID: 31931509 PMCID: PMC7235267 DOI: 10.1038/s41386-020-0611-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 11/09/2022]
Abstract
Nicotine enhances the reinforcement of non-drug rewards by increasing nucleus accumbens (NAcc) reactivity to anticipatory cues. This anticipatory effect is selective as no clear evidence has emerged showing that nicotine acutely changes reward receipt reactivity. However, repeated rewarding experiences shift peak brain reactivity from hedonic reward outcome to the motivational anticipatory cue yielding more habitual cue-induced behavior. Given nicotine's influence on NAcc reactivity and connectivity, it is plausible that nicotine acutely induces this shift and alters NAcc functional connectivity during reward processing. To evaluate this currently untested hypothesis, a randomized crossover design was used in which healthy non-smokers were administered placebo and nicotine (2-mg lozenge). Brain activation to monetary reward anticipation and outcome was evaluated with functional magnetic resonance imaging. Relative to placebo, nicotine induced more NAcc reactivity to reward anticipation. Greater NAcc activation during anticipation was significantly associated with lower NAcc activation to outcome. During outcome, nicotine reduced NAcc functional connectivity with cortical regions including the anterior cingulate cortex, orbitofrontal cortex, and insula. These regions showed the same negative relationship between reward anticipation and outcome as noted in the NAcc. The current findings significantly improve our understanding of how nicotine changes corticostriatal circuit function and communication during distinct phases of reward processing and critically show that these alterations happen acutely following a single dose. The implications of this work explain nicotinic modulation of general reward function, which offer insights into the initial drive to smoke and the subsequent difficulty in cessation.
Collapse
Affiliation(s)
- Kainan S. Wang
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - Maya Zegel
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA
| | - Elena Molokotos
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,0000 0001 0684 8852grid.264352.4Department of Psychology, Suffolk University, Boston, MA USA
| | - Lauren V. Moran
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - David P. Olson
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - Diego A. Pizzagalli
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| | - Amy C. Janes
- 0000 0000 8795 072Xgrid.240206.2McLean Imaging Center, McLean Hospital, Belmont, MA USA ,000000041936754Xgrid.38142.3cHarvard Medical School, Boston, MA USA
| |
Collapse
|
26
|
Haddar M, Uno K, Azuma K, Muramatsu S, Nitta A. Inhibitory effects of Shati/Nat8l overexpression in the medial prefrontal cortex on methamphetamine-induced conditioned place preference in mice. Addict Biol 2020; 25:e12749. [PMID: 30950164 PMCID: PMC7187255 DOI: 10.1111/adb.12749] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/24/2019] [Accepted: 02/28/2019] [Indexed: 11/29/2022]
Abstract
Shati/Nat8l is a novel N-acetyltransferase identified in the brain of mice treated with methamphetamine (METH). Shati/Nat8l mRNA is expressed in various brain areas, including the prefrontal cortex (PFC), where the expression level is higher than that in other brain regions. Shati/Nat8l overexpression in the nucleus accumbens (NAc) attenuates the pharmacological response to METH via mGluR3. Meanwhile, dopamine (DA) and glutamate dysregulations have been reported in the medial prefrontal cortex (mPFC) and NAc after METH self-administration and during reinstatement. However, the mechanism, the reward system, and function of Shati/Nat8l in the mPFC is unclear. Here, we injected an adeno-associated virus (AAV) vector containing Shati/Nat8l into the mPFC of mice, to overexpress Shati/Nat8l in the mPFC (mPFC-Shati/Nat8l). Interestingly, the METH-induced conditioned place preference (CPP) was attenuated in the mPFC-Shati/Nat8l mice, but locomotor activity was not. Additionally, immunohistochemical results from mice that were injected with AAV-GFP showed fluorescence in the mPFC and other brain regions, mainly the NAc, indicating an mPFC-NAc top-down connection. Finally, in vivo microdialysis experiments revealed that Shati/Nat8l overexpression in the mPFC reduced extracellular DA levels and suppressed the METH-induced DA increase in the NAc. Moreover, decreased extracellular glutamate levels were observed in the NAc. These results indicate that Shati/Nat8l overexpression in the mPFC attenuates METH-induced CPP by decreasing extracellular DA in the NAc. In contrast, Shati/Nat8l-mPFC overexpression did not alter METH-induced hyperlocomotion. This study demonstrates that Shati/Nat8l in the mPFC attenuates METH reward-seeking behaviour but not the psychomotor activity of METH.
Collapse
Affiliation(s)
- Meriem Haddar
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical SciencesUniversity of Toyama Toyama Japan
| | - Kyosuke Uno
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical SciencesUniversity of Toyama Toyama Japan
| | - Katsunori Azuma
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical SciencesUniversity of Toyama Toyama Japan
| | - Shin‐ichi Muramatsu
- Division of Neurology, Department of MedicineJichi Medical University Shimotsuke Japan
- Center for Gene and Cell Therapy, The Institute of Medical ScienceThe University of Tokyo Tokyo Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical SciencesUniversity of Toyama Toyama Japan
| |
Collapse
|
27
|
Neural circuits and nicotinic acetylcholine receptors mediate the cholinergic regulation of midbrain dopaminergic neurons and nicotine dependence. Acta Pharmacol Sin 2020; 41:1-9. [PMID: 31554960 PMCID: PMC7468330 DOI: 10.1038/s41401-019-0299-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/06/2019] [Indexed: 12/23/2022]
Abstract
Midbrain dopaminergic (DA) neurons are governed by an endogenous cholinergic system, originated in the mesopontine nuclei. Nicotine hijacks nicotinic acetylcholine receptors (nAChRs) and interferes with physiological function of the cholinergic system. In this review, we describe the anatomical organization of the cholinergic system and the key nAChR subtypes mediating cholinergic regulation of DA transmission and nicotine reward and dependence, in an effort to identify potential targets for smoking intervention. Cholinergic modulation of midbrain DA systems relies on topographic organization of mesopontine cholinergic projections, and activation of nAChRs in midbrain DA neurons. Previous studies have revealed that α4, α6, and β2 subunit-containing nAChRs expressed in midbrain DA neurons and their terminals in the striatum regulate firings of midbrain DA neurons and activity-dependent dopamine release in the striatum. These nAChRs undergo modification upon chronic nicotine exposure. Clinical investigation has demonstrated that partial agonists of these receptors elevate the success rate of smoking cessation relative to placebo. However, further investigations are required to refine the drug targets to mitigate unpleasant side-effects.
Collapse
|
28
|
Mu P, Huang YH. Cholinergic system in sleep regulation of emotion and motivation. Pharmacol Res 2019; 143:113-118. [PMID: 30894329 DOI: 10.1016/j.phrs.2019.03.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/28/2019] [Accepted: 03/15/2019] [Indexed: 01/22/2023]
Abstract
Sleep profoundly regulates our emotional and motivational state of mind. Human brain imaging and animal model studies are providing initial insights on the underlying neural mechanisms. Here, we focus on the brain cholinergic system, including cholinergic neurons in the basal forebrain, ventral striatum, habenula, and brain stem. Although much is learned about cholinergic regulations of emotion and motivation, less is known on their interactions with sleep. Specifically, we present an anatomical framework that highlights cholinergic signaling in the integrated reward-arousal/sleep circuitry, and identify the knowledge gaps on the potential roles of cholinergic system in sleep-mediated regulation of emotion and motivation. Sleep impacts every aspect of brain functions. It not only restores cognitive control, but also retunes emotional and motivational regulation [1]. Sleep disturbance is a comorbidity and sometimes a predicting factor for various psychiatric diseases including major depressive disorder, anxiety, post-traumatic stress disorder, and drug addiction [2-9]. Although it is well recognized that sleep prominently shapes emotional and motivational regulation, the underlying neural mechanisms remain elusive. The brain cholinergic system is essential for a diverse variety of functions including cognition, learning and memory, sensory and motor processing, sleep and arousal, reward processing, and emotion regulation [10-14]. Although cholinergic functions in cognition, learning and memory, motor control, and sleep and arousal have been well established, its interaction with sleep in regulating emotion and motivation has not been extensively studied. Here we review current evidence on sleep-mediated regulation of emotion and motivation, and reveal knowledge gaps on potential contributions from the cholinergic system.
Collapse
Affiliation(s)
- Ping Mu
- College of Life Sciences, Ludong University, 186 Hongqi Middle Road, Yantai, Shandong, 264025, China.
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, 15219, PA, United States.
| |
Collapse
|
29
|
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
|
30
|
Downs AM, Fan X, Donsante C, Jinnah HA, Hess EJ. Trihexyphenidyl rescues the deficit in dopamine neurotransmission in a mouse model of DYT1 dystonia. Neurobiol Dis 2019; 125:115-122. [PMID: 30707939 DOI: 10.1016/j.nbd.2019.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/14/2019] [Accepted: 01/20/2019] [Indexed: 11/17/2022] Open
Abstract
Trihexyphenidyl, a nonselective muscarinic receptor antagonist, is the small molecule drug of choice for the treatment of DYT1 dystonia, but it is poorly tolerated due to significant side effects. A better understanding of the mechanism of action of trihexyphenidyl is needed for the development of improved treatments. Because DTY1 dystonia is associated with both abnormal cholinergic neurotransmission and abnormal dopamine regulation, we tested the hypothesis that trihexyphenidyl normalizes striatal dopamine release in a mouse model of DYT1 dystonia using ex vivo fast scan cyclic voltammetry and in vivo microdialysis. Trihexyphenidyl increased striatal dopamine release and efflux as assessed by ex vivo voltammetry and in vivo microdialysis respectively. In contrast, ʟ-DOPA, which is not usually effective for the treatment of DYT1 dystonia, did not increase dopamine release in either Dyt1 or control mice. Trihexyphenidyl was less effective at enhancing dopamine release in Dyt1 mice relative to controls ex vivo (mean increase WT: 65% vs Dyt1: 35%). Trihexyphenidyl required nicotinic receptors but not glutamate receptors to increase dopamine release. Dyt1 mice were more sensitive to the dopamine release decreasing effects of nicotinic acetylcholine receptor antagonism (IC50: WT = 29.46 nM, Dyt1 = 12.26 nM) and less sensitive to acetylcholinesterase inhibitors suggesting that nicotinic acetylcholine receptor neurotransmission is altered in Dyt1 mice, that nicotinic receptors indirectly mediate the differential effects of trihexyphenidyl in Dyt1 mice, and that nicotinic receptors may be suitable therapeutic targets for DYT1 dystonia.
Collapse
Affiliation(s)
- Anthony M Downs
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Xueliang Fan
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Christine Donsante
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - H A Jinnah
- Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Human Genetics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA
| | - Ellen J Hess
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA.
| |
Collapse
|
31
|
Mansouri-Guilani N, Bernard V, Vigneault E, Vialou V, Daumas S, El Mestikawy S, Gangarossa G. VGLUT3 gates psychomotor effects induced by amphetamine. J Neurochem 2019; 148:779-795. [PMID: 30556914 DOI: 10.1111/jnc.14644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/12/2018] [Accepted: 12/05/2018] [Indexed: 12/26/2022]
Abstract
Several subtypes of modulatory neurons co-express vesicular glutamate transporters (VGLUTs) in addition to their cognate vesicular transporters. These neurons are believed to establish new forms of neuronal communication. The atypical VGLUT3 is of particular interest since in the striatum this subtype is found in tonically active cholinergic interneurons (TANs) and in a subset of 5-HT fibers. The striatum plays a major role in psychomotor effects induced by amphetamine. Whether and how VGLUT3-operated glutamate/ACh or glutamate/5HT co-transmissions modulates psychostimulants-induced maladaptive behaviors is still unknown. Here, we investigate the involvement of VGLUT3 and glutamate co-transmission in amphetamine-induced psychomotor effects and stereotypies. Taking advantage of constitutive and cell-type specific VGLUT3-deficient mouse lines, we tackled the hypothesis that VGLUT3 could gate psychomotor effects (locomotor activity and stereotypies) induced by acute or chronic administration of amphetamine. Interestingly, VGLUT3-null mice demonstrated blunted amphetamine-induced stereotypies as well as reduced striatal ∆FosB expression. VGLUT3-positive varicosities within the striatum arise in part from 5HT neurons. We tested the involvement of VGLUT3 deletion in serotoninergic neurons in amphetamine-induced stereotypies. Mice lacking VGLUT3 specifically in 5HT fibers showed no alteration to amphetamine sensitivity. In contrast, specific deletion of VGLUT3 in cholinergic neurons partially phenocopied the effects observed in the constitutive knock-out mice. Our results show that constitutive deletion of VGLUT3 modulates acute and chronic locomotor effects induced by amphetamine. They point to the fact that the expression of VGLUT3 in multiple brain areas is pivotal in gating amphetamine-induced psychomotor adaptations. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Nina Mansouri-Guilani
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Véronique Bernard
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Erika Vigneault
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Vincent Vialou
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Stéphanie Daumas
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Salah El Mestikawy
- Neuroscience ParisSeine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France.,Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada
| | - Giuseppe Gangarossa
- Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada.,Unité de Biologie Fonctionnelle et Adaptative (BFA) CNRS UMR8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
32
|
Ostroumov A, Dani JA. Inhibitory Plasticity of Mesocorticolimbic Circuits in Addiction and Mental Illness. Trends Neurosci 2018; 41:898-910. [PMID: 30149979 PMCID: PMC6252277 DOI: 10.1016/j.tins.2018.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/12/2018] [Accepted: 07/31/2018] [Indexed: 12/19/2022]
Abstract
Behavioral adaptations occur through remodeling of brain circuits, as arising, for instance, from experience-dependent synaptic plasticity. Drugs of abuse and aversive stimuli, such as stress, act on the mesocorticolimbic system, dysregulating adaptive mechanisms and leading to a variety of aberrant behaviors associated with neuropsychiatric disorders. Until recently, research in the field has commonly focused on experience-dependent synaptic plasticity at excitatory synapses. However, there is growing evidence that synaptic plasticity within inhibitory circuits is an important contributor to maladaptive behaviors. We speculate that restoring normal inhibitory synaptic transmission is a promising therapeutic target for correcting some of the circuit abnormalities underlying neuropsychiatric disorders.
Collapse
Affiliation(s)
- Alexey Ostroumov
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, PA 19104, USA.
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Robertson BD, Al Jaja AS, MacDonald AA, Hiebert NM, Tamjeedi R, Seergobin KN, Schwarz UI, Kim RB, MacDonald PA. SLC6A3 Polymorphism Predisposes to Dopamine Overdose in Parkinson's Disease. Front Neurol 2018; 9:693. [PMID: 30186226 PMCID: PMC6110885 DOI: 10.3389/fneur.2018.00693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022] Open
Abstract
In Parkinson's disease (PD), cognitive functions mediated by brain regions innervated by ventral tegmental area (VTA) worsen with dopamine replacement therapy, whereas processes relying on regions innervated by the substantia nigra pars compacta (SNc) improve. The SLC6A3 gene encodes the dopamine transporter (DAT). The common 9R polymorphism produces higher DAT concentrations and consequently lower baseline dopamine than SLC6A3 wildtype. Whether SLC6A3 genotype modulates the effect of dopaminergic therapy on cognition in PD is not known. We investigated the effect of dopaminergic therapy and SLC6A3 genotype on encoding and recall of abstract images using the Aggie Figures Learning Test in PD patients. Encoding depends upon brain regions innervated by the VTA, whereas recall is mediated by widespread brain regions, a number innervated by the SNc. We found that dopaminergic therapy worsened encoding of abstract images in 9R carriers only. In contrast, dopaminergic therapy improved recall of abstract images in all PD patients, irrespective of SLC6A3 genotype. Our findings suggest that 9R-carrier PD patients are more predisposed to dopamine overdose and medication-induced impairment of cognitive functions mediated by VTA-innervated brain regions. Interestingly, PD patients without the 9R polymorphism did not show such an impairment. SLC6A3 genotype does not modulate the dopaminergic therapy-induced improvement of functions mediated by SNc-innervated regions in PD patients.
Collapse
Affiliation(s)
- Brian D Robertson
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Abdullah S Al Jaja
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada.,Department of Neuroscience, University of Western Ontario, London, ON, Canada
| | - Alex A MacDonald
- Department of Medicine, Undergraduate Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nole M Hiebert
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | | | - Ken N Seergobin
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
| | - Ute I Schwarz
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Richard B Kim
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Penny A MacDonald
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada.,Department of Neuroscience, University of Western Ontario, London, ON, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
34
|
Fortin GM, Ducrot C, Giguère N, Kouwenhoven WM, Bourque MJ, Pacelli C, Varaschin RK, Brill M, Singh S, Wiseman PW, Trudeau LÉ. Segregation of dopamine and glutamate release sites in dopamine neuron axons: regulation by striatal target cells. FASEB J 2018; 33:400-417. [PMID: 30011230 DOI: 10.1096/fj.201800713rr] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Dopamine (DA) is a key regulator of circuits controlling movement and motivation. A subset of midbrain DA neurons has been shown to express the vesicular glutamate transporter (VGLUT)2, underlying their capacity for glutamate release. Glutamate release is found mainly by DA neurons of the ventral tegmental area (VTA) and can be detected at terminals contacting ventral, but not dorsal, striatal neurons, suggesting the possibility that target-derived signals regulate the neurotransmitter phenotype of DA neurons. Whether glutamate can be released from the same terminals that release DA or from a special subset of axon terminals is unclear. Here, we provide in vitro and in vivo data supporting the hypothesis that DA and glutamate-releasing terminals in mice are mostly segregated and that striatal neurons regulate the cophenotype of midbrain DA neurons and the segregation of release sites. Our work unveils a fundamental feature of dual neurotransmission and plasticity of the DA system.-Fortin, G. M., Ducrot, C., Giguère, N., Kouwenhoven, W. M., Bourque, M.-J., Pacelli, C., Varaschin, R. K., Brill, M., Singh, S., Wiseman, P. W., Trudeau, L.-E. Segregation of dopamine and glutamate release sites in dopamine neuron axons: regulation by striatal target cells.
Collapse
Affiliation(s)
- Guillaume M Fortin
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | - Charles Ducrot
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas Giguère
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| | | | - Marie-Josée Bourque
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | - Consiglia Pacelli
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | | | - Marion Brill
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada
| | - Sherdeep Singh
- Department of Chemistry, McGill University, Montreal, Quebec, Canada; and
| | - Paul W Wiseman
- Department of Chemistry, McGill University, Montreal, Quebec, Canada; and
| | - Louis-Éric Trudeau
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Quebec, Canada.,Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Vo A, Seergobin KN, MacDonald PA. Independent effects of age and levodopa on reversal learning in healthy volunteers. Neurobiol Aging 2018; 69:129-139. [PMID: 29894903 DOI: 10.1016/j.neurobiolaging.2018.05.014] [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: 06/29/2017] [Revised: 05/02/2018] [Accepted: 05/10/2018] [Indexed: 11/25/2022]
Abstract
The dopamine overdose hypothesis has provided an important theoretical framework for understanding cognition in Parkinson's disease. It posits that effects of dopaminergic therapy on cognition in Parkinson's disease depend on baseline dopamine levels in brain regions that support different functions. Although functions performed by more severely dopamine-depleted brain regions improve with medication, those associated with less dopamine deficient areas are actually worsened. It is presumed that medication-related worsening of cognition owes to dopamine overdose. We investigated whether age-related changes in baseline dopamine levels would modulate effects of dopaminergic therapy on reward learning in healthy volunteers. In a double-blind, crossover design, healthy younger and older adults completed a probabilistic reversal learning task after treatment with 100/25 mg of levodopa/carbidopa versus placebo. Older adults learned more poorly than younger adults at baseline, being more likely to shift responses after misleading punishment. Levodopa worsened stimulus-reward learning relative to placebo to the same extent in both groups, irrespective of differences in baseline performance and expected dopamine levels. When order effects were eliminated, levodopa induced response shifts after reward more often than placebo. Our results reveal independent deleterious effects of age group and exogenous dopamine on reward learning, suggesting a more complex scenario than predicted by the dopamine overdose hypothesis.
Collapse
Affiliation(s)
- Andrew Vo
- Brain and Mind Institute, University of Western Ontario, London, Canada; Department of Psychology, University of Western Ontario, London, Canada
| | - Ken N Seergobin
- Brain and Mind Institute, University of Western Ontario, London, Canada
| | - Penny A MacDonald
- Brain and Mind Institute, University of Western Ontario, London, Canada; Department of Psychology, University of Western Ontario, London, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.
| |
Collapse
|
38
|
Region-Specific Regulation of Presynaptic Dopamine Homeostasis by D 2 Autoreceptors Shapes the In Vivo Impact of the Neuropsychiatric Disease-Associated DAT Variant Val559. J Neurosci 2018; 38:5302-5312. [PMID: 29739866 DOI: 10.1523/jneurosci.0055-18.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/19/2018] [Accepted: 04/14/2018] [Indexed: 12/21/2022] Open
Abstract
Disruptions of dopamine (DA) signaling contribute to a broad spectrum of neuropsychiatric disorders, including attention-deficit hyperactivity disorder (ADHD), addiction, bipolar disorder, and schizophrenia. Despite evidence that risk for these disorders derives from heritable variation in DA-linked genes, a better understanding is needed of the molecular and circuit context through which gene variation drives distinct disease traits. Previously, we identified the DA transporter (DAT) variant Val559 in subjects with ADHD and established that the mutation supports anomalous DAT-mediated DA efflux (ADE). Here, we demonstrate that region-specific contributions of D2 autoreceptors (D2AR) to presynaptic DA homeostasis dictate the consequences of Val559 expression in adolescent male mice. We show that activation of D2ARs in the WT dorsal striatum (DS), but not ventral striatum (VS), increases DAT phosphorylation and surface trafficking. In contrast, the activity of tyrosine hydroxylase (TH) is D2AR-dependent in both regions. In the DS but not VS of Val559 mice, tonic activation of D2ARs drives a positive feedback loop that promotes surface expression of efflux-prone DATs, raising extracellular DA levels and overwhelming DAT-mediated DA clearance capacity. Whereas D2ARs that regulate DAT are tonically activated in the Val559 DS, D2ARs that regulate TH become desensitized, allowing maintenance of cytosolic DA needed to sustain ADE. Together with prior findings, our results argue for distinct D2AR pools that regulate DA synthesis versus DA release and inactivation and offer a clear example of how the penetrance of gene variation can be limited to a subset of expression sites based on differences in intersecting regulatory networks.SIGNIFICANCE STATEMENT Altered dopamine (DA) signaling has been linked to multiple neuropsychiatric disorders. In an effort to understand and model disease-associated DAergic disturbances, we previously screened the DA transporter (DAT) in subjects with attention-deficit hyperactivity disorder (ADHD) and identified multiple, functionally impactful, coding variants. One of these variants, Val559, supports anomalous DA efflux (ADE) and in transgenic mice leads to changes in locomotor patterns, psychostimulant sensitivity, and impulsivity. Here, we show that the penetrance of Val559 ADE is dictated by region-specific differences in how presynaptic D2-type autoreceptors (D2ARs) constrain DA signaling, biasing phenotypic effects to dorsal striatal projections. The Val559 model illustrates how the impact of genetic variation underlying neuropsychiatric disorders can be shaped by the differential engagement of synaptic regulatory mechanisms.
Collapse
|
39
|
Chen YH, Kuo TT, Yi-Kung Huang E, Chou YC, Chiang YH, Hoffer BJ, Miller J. Effect of traumatic brain injury on nicotine-induced modulation of dopamine release in the striatum and nucleus accumbens shell. Oncotarget 2018. [PMID: 29515787 PMCID: PMC5839368 DOI: 10.18632/oncotarget.24245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Traumatic brain injury is associated with substantial alterations in reward processing, but underlying mechanisms are controversial. Objective A better understanding of alterations in dopamine (DA) release patterns from the dorsal striatum and nucleus accumbens shell (NAc) may provide insights into posttraumatic reward pathology. Materials and Methods The patterns of DA release with or without exposure to nicotine in brain slices with striatum and NAc, isolated from Sprague-Dawley rats with 6 psi fluid percussion (FPI) or sham injury were analysis by using fast-scan cyclic voltammetry. Tonic and phasic DA releases were assessed using single pulse and 10 pulses at 25 Hz, respectively. DA release relative to stimulation intensity, frequency, number of pulses, and paired-pulse facilitation was evaluated to determine release probability and response to bursting. Results There was a profound suppression in tonic DA release after nicotine desensitization after FPI, and the input/output curve for the DA release based on stimulation intensity was shifted to the right. FPI was associated with a significant decrease in frequency-dependent DA release augmentation, DA release induced by high frequency stimulation trains, and DA release in response to paired-pulse facilitation. The effect of nicotine desensitization was similar in FPI and sham-injured animals, although significantly smaller after FPI. Nicotine desensitization–induced differences between phasic and tonic release concentrations that contrasted with the reward-related signals then became less prominent in NAc after FPI. Conclusions TBI blunts DA release from mesolimbic reward centers, and more intense stimuli are required to produce context-dependent DA release sufficient to have a physiological effect. Implications The nicotine desensitization-related suppression in tonic DA release was profound with right-ward shift of the input/output curve for DA release after FPI. FPI was associated with a significant decrease in frequency-dependent DA release augmentation, DA release induced by high frequency stimulation trains, and DA release in response to paired-pulse facilitation. Nicotine desensitization–induced differences between phasic and tonic release concentrations that contrasted with the reward-related signals then became less prominent in NAc after FPI. TBI thus blunts DA release from mesolimbic reward centers, and more intense stimuli are required to produce context-dependent DA release sufficient to have a physiological effect.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Tung-Tai Kuo
- Graduate Institute of Computer and Communication Engineering, National Taipei University of Technology, Taipei, Taiwan, R.O.C
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Yu-Ching Chou
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Yung-Hsiao Chiang
- Graduate Program on Neuroregeneration, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Barry J Hoffer
- Graduate Program on Neuroregeneration, Taipei Medical University, Taipei, Taiwan, R.O.C.,Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jonathon Miller
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
40
|
Beloate LN, Coolen LM. Influences of social reward experience on behavioral responses to drugs of abuse: Review of shared and divergent neural plasticity mechanisms for sexual reward and drugs of abuse. Neurosci Biobehav Rev 2017; 83:356-372. [DOI: 10.1016/j.neubiorev.2017.10.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 10/25/2022]
|
41
|
Chiamulera C, Marzo CM, Balfour DJK. Metabotropic glutamate receptor 5 as a potential target for smoking cessation. Psychopharmacology (Berl) 2017; 234:1357-1370. [PMID: 27847973 DOI: 10.1007/s00213-016-4487-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/07/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Most habitual smokers find it difficult to quit smoking because they are dependent upon the nicotine present in tobacco smoke. Tobacco dependence is commonly treated pharmacologically using nicotine replacement therapy or drugs, such as varenicline, that target the nicotinic receptor. Relapse rates, however, remain high, and there remains a need to develop novel non-nicotinic pharmacotherapies for the dependence that are more effective than existing treatments. OBJECTIVE The purpose of this paper is to review the evidence from preclinical and clinical studies that drugs that antagonise the metabotropic glutamate receptor 5 (mGluR5) in the brain are likely to be efficacious as treatments for tobacco dependence. RESULTS Imaging studies reveal that chronic exposure to tobacco smoke reduces the density of mGluR5s in human brain. Preclinical results demonstrate that negative allosteric modulators (NAMs) at mGluR5 attenuate both nicotine self-administration and the reinstatement of responding evoked by exposure to conditioned cues paired with nicotine delivery. They also attenuate the effects of nicotine on brain dopamine pathways implicated in addiction. CONCLUSIONS Although mGluR5 NAMs attenuate most of the key facets of nicotine dependence, they potentiate the symptoms of nicotine withdrawal. This may limit their value as smoking cessation aids. The NAMs that have been employed most widely in preclinical studies of nicotine dependence have too many "off-target" effects to be used clinically. However, newer mGluR5 NAMs have been developed for clinical use in other indications. Future studies will determine if these agents can also be used effectively and safely to treat tobacco dependence.
Collapse
Affiliation(s)
- Cristiano Chiamulera
- Neuropsychopharmacology Lab., Section Pharmacology, Department Diagnostic and Public Health, University of Verona, P.le Scuro 10, 37134, Verona, Italy.
| | - Claudio Marcello Marzo
- Neuropsychopharmacology Lab., Section Pharmacology, Department Diagnostic and Public Health, University of Verona, P.le Scuro 10, 37134, Verona, Italy
| | - David J K Balfour
- Division of Neuroscience, University of Dundee Medical School, Mailbox 6, Ninewells Hospital, Dundee, DD1 9SY, UK
| |
Collapse
|
42
|
Lv X, Dickerson JW, Rook JM, Lindsley CW, Conn PJ, Xiang Z. M 1 muscarinic activation induces long-lasting increase in intrinsic excitability of striatal projection neurons. Neuropharmacology 2017; 118:209-222. [PMID: 28336323 DOI: 10.1016/j.neuropharm.2017.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 02/20/2017] [Accepted: 03/15/2017] [Indexed: 01/11/2023]
Abstract
The dorsolateral striatum is critically involved in movement control and motor learning. Striatal function is regulated by a variety of neuromodulators including acetylcholine. Previous studies have shown that cholinergic activation excites striatal principal projection neurons, medium spiny neurons (MSNs), and this action is mediated by muscarinic acetylcholine subtype 1 receptors (M1) through modulating multiple potassium channels. In the present study, we used electrophysiology techniques in conjunction with optogenetic and pharmacological tools to determine the long-term effects of striatal cholinergic activation on MSN intrinsic excitability. A transient increase in acetylcholine release in the striatum by optogenetic stimulation resulted in a long-lasting increase in excitability of MSNs, which was associated with hyperpolarizing shift of action potential threshold and decrease in afterhyperpolarization (AHP) amplitude, leading to an increase in probability of EPSP-action potential coupling. The M1 selective antagonist VU0255035 prevented, while the M1 selective positive allosteric modulator (PAM) VU0453595 potentiated the cholinergic activation-induced persistent increase in MSN intrinsic excitability, suggesting that M1 receptors are critically involved in the induction of this long-lasting response. This M1 receptor-dependent long-lasting change in MSN intrinsic excitability could have significant impact on striatal processing and might provide a novel mechanism underlying cholinergic regulation of the striatum-dependent motor learning and cognitive function. Consistent with this, behavioral studies indicate that potentiation of M1 receptor signaling by VU0453595 enhanced performance of mice in cue-dependent water-based T-maze, a dorsolateral striatum-dependent learning task.
Collapse
Affiliation(s)
- Xiaohui Lv
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jonathan W Dickerson
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jerri M Rook
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
43
|
Gomez-A A, Fiorenza AM, Boschen SL, Sugi AH, Beckman D, Ferreira ST, Lee K, Blaha CD, Da Cunha C. Diazepam Inhibits Electrically Evoked and Tonic Dopamine Release in the Nucleus Accumbens and Reverses the Effect of Amphetamine. ACS Chem Neurosci 2017; 8:300-309. [PMID: 28038309 DOI: 10.1021/acschemneuro.6b00358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diazepam is a benzodiazepine receptor agonist with anxiolytic and addictive properties. Although most drugs of abuse increase the level of release of dopamine in the nucleus accumbens, here we show that diazepam not only causes the opposite effect but also prevents amphetamine from enhancing dopamine release. We used 20 min sampling in vivo microdialysis and subsecond fast-scan cyclic voltammetry recordings at carbon-fiber microelectrodes to show that diazepam caused a dose-dependent decrease in the level of tonic and electrically evoked dopamine release in the nucleus accumbens of urethane-anesthetized adult male Swiss mice. In fast-scan cyclic voltammetry assays, dopamine release was evoked by electrical stimulation of the ventral tegmental area. We observed that 2 and 3 mg of diazepam/kg reduced the level of electrically evoked dopamine release, and this effect was reversed by administration of the benzodiazepine receptor antagonist flumazenil in doses of 2.5 and 5 mg/kg, respectively. No significant effects on measures of dopamine re-uptake were observed. Cyclic voltammetry experiments further showed that amphetamine (5 mg/kg, intraperitoneally) caused a significant increase in the level of dopamine release and in the half-life for dopamine re-uptake. Diazepam (2 mg/kg) significantly weakened the effect of amphetamine on dopamine release without affecting dopamine re-uptake. These results suggest that the pharmacological effects of benzodiazepines have a dopaminergic component. In addition, our findings challenge the classic view that all drugs of abuse cause dopamine release in the nucleus accumbens and suggest that benzodiazepines could be useful in the treatment of addiction to other drugs that increase the level of dopamine release, such as cocaine, amphetamines, and nicotine.
Collapse
Affiliation(s)
- Alexander Gomez-A
- Departamento
de Farmacologia, Universidade Federal do Paraná, Curitiba 81.530-980, PR, Brazil
| | - Amanda M. Fiorenza
- Departamento
de Farmacologia, Universidade Federal do Paraná, Curitiba 81.530-980, PR, Brazil
| | - Suelen L. Boschen
- Departamento
de Farmacologia, Universidade Federal do Paraná, Curitiba 81.530-980, PR, Brazil
- Institute
of Biophysics Carlos Chagas Filho and Institute of Medical Biochemistry
Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de
Janeiro, Brazil
| | - Adam H. Sugi
- Departamento
de Farmacologia, Universidade Federal do Paraná, Curitiba 81.530-980, PR, Brazil
| | - Danielle Beckman
- Institute
of Biophysics Carlos Chagas Filho and Institute of Medical Biochemistry
Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de
Janeiro, Brazil
| | - Sergio T. Ferreira
- Institute
of Biophysics Carlos Chagas Filho and Institute of Medical Biochemistry
Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de
Janeiro, Brazil
| | - Kendall Lee
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Charles D. Blaha
- Department
of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Claudio Da Cunha
- Departamento
de Farmacologia, Universidade Federal do Paraná, Curitiba 81.530-980, PR, Brazil
| |
Collapse
|
44
|
Criscitelli K, Avena NM. The neurobiological and behavioral overlaps of nicotine and food addiction. Prev Med 2016; 92:82-89. [PMID: 27509870 DOI: 10.1016/j.ypmed.2016.08.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 08/01/2016] [Accepted: 08/06/2016] [Indexed: 12/19/2022]
Abstract
Both cigarette smoking and obesity are significant public health concerns and are associated with increased risk of early mortality. It is well established that the mesolimbic dopamine pathway is an important component of the reward system within the brain and is implicated in the development of addiction. Indeed, nicotine and highly palatable foods are capable of altering dopamine release within this system, engendering addictive like responses in susceptible individuals. Although additional research is warranted, findings from animal and human literature have elucidated many of neuroadaptions that occur from exposure to nicotine and highly palatable foods, leading to a greater understanding of the underlying mechanisms contributing to these aberrant behaviors. In this review we present the findings taken from preclinical and clinical literature of the known effects of exposure to nicotine and highly palatable foods on the reward related circuitry within the brain. Further, we compare the neurobiological and behavioral overlaps between nicotine, highly palatable foods and obesity. Lastly, we examine the stigma associated with smoking, obesity and food addiction, and the consequences stigma has on the overall health and wellbeing of an individual.
Collapse
Affiliation(s)
- Kristen Criscitelli
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicole M Avena
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
45
|
Salinas AG, Davis MI, Lovinger DM, Mateo Y. Dopamine dynamics and cocaine sensitivity differ between striosome and matrix compartments of the striatum. Neuropharmacology 2016; 108:275-83. [PMID: 27036891 PMCID: PMC5026225 DOI: 10.1016/j.neuropharm.2016.03.049] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/08/2016] [Accepted: 03/28/2016] [Indexed: 12/13/2022]
Abstract
The striatum is typically classified according to its major output pathways, which consist of dopamine D1 and D2 receptor-expressing neurons. The striatum is also divided into striosome and matrix compartments, based on the differential expression of a number of proteins, including the mu opioid receptor, dopamine transporter (DAT), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1). Numerous functional differences between the striosome and matrix compartments are implicated in dopamine-related neurological disorders including Parkinson's disease and addiction. Using Nr4a1-eGFP mice, we provide evidence that electrically evoked dopamine release differs between the striosome and matrix compartments in a regionally-distinct manner. We further demonstrate that this difference is not due to differences in inhibition of dopamine release by dopamine autoreceptors or nicotinic acetylcholine receptors. Furthermore, cocaine enhanced extracellular dopamine in striosomes to a greater degree than in the matrix and concomitantly inhibited dopamine uptake in the matrix to a greater degree than in striosomes. Importantly, these compartment differences in cocaine sensitivity were limited to the dorsal striatum. These findings demonstrate a level of exquisite microanatomical regulation of dopamine by the DAT in striosomes relative to the matrix.
Collapse
Affiliation(s)
- Armando G Salinas
- The Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA; Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD 20852, USA
| | - Margaret I Davis
- Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD 20852, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD 20852, USA
| | - Yolanda Mateo
- Laboratory for Integrative Neuroscience, Section on Synaptic Pharmacology, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD 20852, USA.
| |
Collapse
|
46
|
Falcone M, Cao W, Bernardo L, Tyndale RF, Loughead J, Lerman C. Brain Responses to Smoking Cues Differ Based on Nicotine Metabolism Rate. Biol Psychiatry 2016; 80:190-7. [PMID: 26805583 PMCID: PMC5625335 DOI: 10.1016/j.biopsych.2015.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/22/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inherited differences in the rate of metabolism of nicotine, the addictive chemical in tobacco, affect smoking behavior and quitting success. The nicotine metabolite ratio (3'-hydroxycotinine/cotinine) is a reliable measure of nicotine clearance and a well-validated predictive biomarker of response to pharmacotherapy. To clarify the mechanisms underlying these associations, we investigated the neural responses to smoking cues in normal and slow nicotine metabolizers. METHODS Treatment-seeking smokers (N = 69; 30 slow metabolizers and 39 normal metabolizers) completed a visual cue reactivity task during functional magnetic resonance imaging on two separate occasions: once during smoking satiety and once after 24 hours of smoking abstinence. RESULTS In whole-brain analysis, normal (compared with slow) metabolizers exhibited heightened abstinence-induced neural responses to smoking cues in the left caudate, left inferior frontal gyrus, and left frontal pole. These effects were more pronounced when extreme groups of slow and normal metabolizers were examined. Greater activation in the left caudate and left frontal pole was associated with abstinence-induced subjective cravings to smoke. CONCLUSIONS Inherited differences in rate of nicotine elimination may drive neural responses to smoking cues during early abstinence, providing a plausible mechanism to explain differences in smoking behaviors and response to cessation treatment. Normal metabolizers may benefit from adjunctive behavioral smoking cessation treatments, such as cue exposure therapy.
Collapse
Affiliation(s)
- Mary Falcone
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, 3535 Market Street, Suite 4100, Philadelphia, PA 19104
| | - Wen Cao
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, 3535 Market Street, Suite 4100, Philadelphia, PA 19104
| | - Leah Bernardo
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, 3535 Market Street, Suite 4100, Philadelphia, PA 19104
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Departments of Psychiatry, and Pharmacology & Toxicology, University of Toronto, Canada M5S 1A8
| | - James Loughead
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, 3535 Market Street, Suite 4100, Philadelphia, PA 19104
| | - Caryn Lerman
- Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
47
|
Rademacher L, Prinz S, Winz O, Henkel K, Dietrich CA, Schmaljohann J, Mohammadkhani Shali S, Schabram I, Stoppe C, Cumming P, Hilgers RD, Kumakura Y, Coburn M, Mottaghy FM, Gründer G, Vernaleken I. Effects of Smoking Cessation on Presynaptic Dopamine Function of Addicted Male Smokers. Biol Psychiatry 2016; 80:198-206. [PMID: 26803340 DOI: 10.1016/j.biopsych.2015.11.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/25/2015] [Accepted: 11/11/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND There is evidence of abnormal cerebral dopamine transmission in nicotine-dependent smokers, but it is unclear whether dopaminergic abnormalities are due to acute nicotine abuse or whether they persist with abstinence. We addressed this question by conducting longitudinal positron emission tomography (PET) examination of smokers before and after 3 months of abstinence. METHODS We obtained baseline 6-[(18)F]fluoro-L-DOPA (FDOPA)-PET scans in 15 nonsmokers and 30 nicotine-dependent smokers, who either smoked as per their usual habit or were in acute withdrawal. All smokers then underwent cessation treatment, and successful abstainers were re-examined by FDOPA-PET after 3 months of abstinence (n = 15). Uptake of FDOPA was analyzed using a steady-state model yielding estimates of the dopamine synthesis capacity (K); the turnover of tracer dopamine formed in living brain (kloss); and the tracer distribution volume (Vd), which is an index of dopamine storage capacity. RESULTS Compared with nonsmokers, K was 15% to 20% lower in the caudate nuclei of consuming smokers. Intraindividual comparisons of consumption and long-term abstinence revealed significant increases in K in the right dorsal and left ventral caudate nuclei. Relative to acute withdrawal, Vd significantly decreased in the right ventral and dorsal caudate after prolonged abstinence. Severity of nicotine dependence significantly correlated with dopamine synthesis capacity and dopamine turnover in the bilateral ventral putamen of consuming smokers. CONCLUSIONS The results suggest a lower dopamine synthesis capacity in nicotine-dependent smokers that appears to normalize with abstinence. Further investigations are needed to clarify the role of dopamine in nicotine addiction to help develop smoking prevention and cessation treatments.
Collapse
Affiliation(s)
- Lena Rademacher
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen; Department of Psychiatry and Psychotherapy, Social Neuroscience Laboratory, University of Lübeck, Lübeck, Germany.
| | - Susanne Prinz
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen; Department of Psychiatry and Psychotherapy, Centre for Integrative Psychiatry, University of Zürich, Rheinau, Switzerland
| | - Oliver Winz
- Department of Nuclear Medicine, RWTH Aachen University, Aachen
| | - Karsten Henkel
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen
| | - Claudia A Dietrich
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen
| | | | | | - Ina Schabram
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen
| | - Christian Stoppe
- Department of Anesthesiology, RWTH Aachen University, Aachen; Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen
| | - Paul Cumming
- Department of Neuropsychiatry and Psychosomatic Medicine, Oslo University Hospital, Oslo, Norway; School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Yoshitaka Kumakura
- Department of Pharmacology and Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University, Aachen
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University, Aachen; Jülich/Aachen Research Alliance, Aachen, Germany; Department of Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Gerhard Gründer
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen; Jülich/Aachen Research Alliance, Aachen, Germany
| | - Ingo Vernaleken
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen; Jülich/Aachen Research Alliance, Aachen, Germany
| |
Collapse
|
48
|
Pérez-Rubio G, Pérez-Rodríguez ME, Fernández-López JC, Ramírez-Venegas A, García-Colunga J, Ávila-Moreno F, Camarena A, Sansores RH, Falfán-Valencia R. SNPs in NRXN1 and CHRNA5 are associated to smoking and regulation of GABAergic and glutamatergic pathways. Pharmacogenomics 2016; 17:1145-1158. [PMID: 27355804 DOI: 10.2217/pgs-2016-0020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
AIM To identify genetic variants associated with greater tobacco consumption in a Mexican population. PATIENTS & METHODS Daily smokers were classified as light smokers (LS; n = 742), heavy smokers (HS; n = 601) and nonsmokers (NS; n = 606). In the first stage, a genotyping microarray that included 347 SNPs in CHRNA2-CHRNA7/CHRNA10, CHRNB2-CHRNB4 and NRXN1 genes and 37 ancestry-informative markers was used to analyze 707 samples (187 HS, 328 LS and 192 NS). In the second stage, 14 SNPs from stage 1 were validated in the remaining samples (HS, LS and NS; n = 414 in each group) using real-time PCR. To predict the role of the associated SNPs, an in silico analysis was performed. RESULTS Two SNPs in NRXN1 and two in CHRNA5 were associated with cigarette consumption, while rs10865246/C (NRXN1) was associated with high nicotine addiction. The in silico analysis revealed that rs1882296/T had a high level of homology with Hsa-miR-6740-5p, which encodes a putative miRNA that targets glutamate receptor subunits (GRIA2, GRID2) and GABA receptor subunits (GABRG1, GABRA4, GABRB2), while rs1882296/C had a high level of homology with Hsa-miR-6866-5p, which encodes a different miRNA that targets GRID2 and GABRB2. CONCLUSION In a Mexican Mestizo population, greater consumption of cigarettes was influenced by polymorphisms in the NRXN1 and CHRNA5 genes. We proposed new hypotheses regarding the putative roles of miRNAs that influence the GABAergic and glutamatergic pathways in smoking addiction.
Collapse
Affiliation(s)
- Gloria Pérez-Rubio
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Laboratorio HLA, México, DF, México.,Instituto Mexicano del Seguro Social., Unidad de Investigación Médica en Inmunología, CMN S-XXI, México, DF, México.,Universidad Nacional Autónoma de México, Posgrado en Ciencias Biológicas, México, DF, México
| | - Martha E Pérez-Rodríguez
- Instituto Mexicano del Seguro Social., Unidad de Investigación Médica en Inmunología, CMN S-XXI, México, DF, México
| | | | - Alejandra Ramírez-Venegas
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Departamento de Investigación en Tabaquismo y EPOC, México, DF, México
| | - Jesús García-Colunga
- Universidad Nacional Autónoma de México, Campus Juriquilla, Instituto de Neurobiología, Departamento de Neurobiología Celular y Molecular, Juriquilla, Querétaro, Mexico
| | - Federico Ávila-Moreno
- Universidad Nacional Autónoma de México, Facultad de Estudios Superiores (FES)-Iztacala, Biomedicine Research Unit (UBIMED), Cancer Epigenomics Laboratory 12, Tlalnepantla, México, México
| | - Angel Camarena
- Universidad Nacional Autónoma de México, Posgrado en Ciencias Biológicas, México, DF, México
| | - Raúl H Sansores
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Departamento de Investigación en Tabaquismo y EPOC, México, DF, México
| | - Ramcés Falfán-Valencia
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Laboratorio HLA, México, DF, México
| |
Collapse
|
49
|
Dobbs LK, Kaplan AR, Lemos JC, Matsui A, Rubinstein M, Alvarez VA. Dopamine Regulation of Lateral Inhibition between Striatal Neurons Gates the Stimulant Actions of Cocaine. Neuron 2016; 90:1100-13. [PMID: 27181061 DOI: 10.1016/j.neuron.2016.04.031] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/11/2016] [Accepted: 04/13/2016] [Indexed: 12/29/2022]
Abstract
Striatal medium spiny neurons (MSNs) form inhibitory synapses on neighboring striatal neurons through axon collaterals. The functional relevance of this lateral inhibition and its regulation by dopamine remains elusive. We show that synchronized stimulation of collateral transmission from multiple indirect-pathway MSNs (iMSNs) potently inhibits action potentials in direct-pathway MSNs (dMSNs) in the nucleus accumbens. Dopamine D2 receptors (D2Rs) suppress lateral inhibition from iMSNs to disinhibit dMSNs, which are known to facilitate locomotion. Surprisingly, D2R inhibition of synaptic transmission was larger at axon collaterals from iMSNs than their projections to the ventral pallidum. Targeted deletion of D2Rs from iMSNs impaired cocaine's ability to suppress lateral inhibition and increase locomotion. These impairments were rescued by chemogenetic activation of Gi-signaling in iMSNs. These findings shed light on the functional significance of lateral inhibition between MSNs and offer a novel synaptic mechanism by which dopamine gates locomotion and cocaine exerts its canonical stimulant response. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Lauren K Dobbs
- Section on Neuronal Structure, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Alanna R Kaplan
- Section on Neuronal Structure, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Julia C Lemos
- Section on Neuronal Structure, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Aya Matsui
- Section on Neuronal Structure, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, C1428ADN, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428ADN, Argentina; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Veronica A Alvarez
- Section on Neuronal Structure, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Harun R, Hare KM, Brough EM, Munoz MJ, Grassi CM, Torres GE, Grace AA, Wagner AK. Fast-scan cyclic voltammetry demonstrates that L-DOPA produces dose-dependent, regionally selective bimodal effects on striatal dopamine kinetics in vivo. J Neurochem 2016; 136:1270-1283. [PMID: 26611352 PMCID: PMC4884169 DOI: 10.1111/jnc.13444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a debilitating condition that is caused by a relatively specific degeneration of dopaminergic (DAergic) neurons of the substantia nigra pars compacta. L-DOPA was introduced as a viable treatment option for PD over 40 years ago and still remains the most common and effective therapy for PD. Though the effects of L-DOPA to augment striatal DA production are well known, little is actually known about how L-DOPA alters the kinetics of DA neurotransmission that contribute to its beneficial and adverse effects. In this study, we examined the effects of L-DOPA administration (50 mg/kg carbidopa + 0, 100, and 250 mg/kg L-DOPA) on regional electrically stimulated DA response kinetics using fast-scan cyclic voltammetry in anesthetized rats. We demonstrate that L-DOPA enhances DA release in both the dorsal striatum (D-STR) and nucleus accumbens (NAc), but surprisingly causes a delayed inhibition of release in the D-STR. In both regions, L-DOPA progressively attenuated reuptake kinetics, predominantly through a decrease in Vmax . These findings have important implications on understanding the pharmacodynamics of L-DOPA, which may be informative for understanding its therapeutic effects and also common side effects like L-DOPA-induced dyskinesias (LID). L-DOPA is commonly used to treat Parkinsonian symptoms, but little is known about how it affects presynaptic DA neurotransmission. Using in vivo fast-scan cyclic voltammetry, we show L-DOPA inhibits DA reuptake in a region-specific and dose-dependent manner, and L-DOPA has paradoxical effects on release. These findings may be important when considering mechanisms for L-DOPA's therapeutic benefits and adverse side-effects.
Collapse
Affiliation(s)
- Rashed Harun
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
| | - Kristin M Hare
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elizabeth M Brough
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
| | - Miranda J Munoz
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biological Sciences, Carnegie Mellon University, Mellon College of Science, Pittsburgh, Pennsylvania, USA
| | - Christine M Grassi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gonzalo E Torres
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Departments of Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy K Wagner
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|