1
|
McLaurin KA, Ott RK, Mactutus CF, Booze RM. Adolescent oral oxycodone self-administration disrupts neurobehavioral and neurocognitive development. Neuropharmacology 2024; 258:110064. [PMID: 38981578 PMCID: PMC11418068 DOI: 10.1016/j.neuropharm.2024.110064] [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: 02/22/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/11/2024]
Abstract
Nonmedical use of prescription opioids peaks during late adolescence, a developmental period associated with the maturation of higher-order cognitive processes. To date, however, how chronic adolescent oxycodone (OXY) self-administration alters neurobehavioral (i.e., locomotion, startle reactivity) and/or neurocognitive (i.e., preattentive processes, intrasession habituation, stimulus-reinforcement learning, sustained attention) function has not yet been systematically evaluated. Hence, the rationale was built for establishing the dose-dependency of adolescent OXY self-administration on the trajectory of neurobehavioral and neurocognitive development. From postnatal day (PD) 35 to PD 105, an age in rats that corresponds to the adolescent and young adult period in humans, male and female F344/N rats received access to either oral OXY (0, 2, 5, or 10 mg/kg) or water under a two-bottle choice experimental paradigm. Independent of biological sex or dose, rodents voluntarily escalated their OXY intake across ten weeks. A longitudinal experimental design revealed prominent OXY-induced impairments in neurobehavioral development, characterized by dose-dependent increases in locomotion and sex-dependent increases in startle reactivity. Systematic manipulation of the interstimulus interval in prepulse inhibition supports an OXY-induced impairment in preattentive processes. Despite the long-term cessation of OXY intake, rodents with a history of chronic adolescent oral OXY self-administration exhibited deficits in sustained attention; albeit no alterations in stimulus-reinforcement learning were observed. Taken together, adolescent oral OXY self-administration induces selective long-term alterations in neurobehavioral and neurocognitive development enjoining the implementation of safer prescribing guidelines for this population.
Collapse
Affiliation(s)
- Kristen A McLaurin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40508, USA.
| | - Rachael K Ott
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Charles F Mactutus
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| | - Rosemarie M Booze
- Cognitive and Neural Science Program, Department of Psychology, Barnwell College, University of South Carolina, 1512 Pendleton Street, Columbia, SC, 29208, USA
| |
Collapse
|
2
|
Randall CA, Sun D, Randall PA. A novel alcohol+nicotine co-use self-administration procedure reveals sex differences and differential alteration of mesocorticolimbic TLR- and cholinergic-related neuroimmune gene expression in rats. Alcohol 2024; 121:115-131. [PMID: 39197504 DOI: 10.1016/j.alcohol.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Although alcohol and nicotine are two of the most commonly co-used drugs with upwards of 90% of adults with an alcohol use disorder (AUD) in the US also smoking, we don't tend to study alcohol and nicotine use this way. The current studies sought to develop and assess a novel alcohol + nicotine co-access self-administration (SA) model in adult male and female Long-Evans rats. Further, both drugs are implicated in neuroimmune function, albeit in largely opposing ways. Chronic alcohol use increases neuroinflammation via toll-like receptors (TLRs) which in turn increases alcohol intake. By contrast, nicotine produces anti-inflammatory effects, in part, through the monomeric alpha7 receptor (ChRNa7). Following long-term co-access (6 months), rats reliably administered both drugs during daily sessions, however males generally responded for more alcohol and females for nicotine. This was reflected in plasma analysis with translationally relevant intake levels of both alcohol and nicotine, making it invaluable in studying the effects of co-use on behavior and CNS function. Moreover, male rats show sensitivity to alterations in alcohol concentration whereas females show sensitivity to alterations in nicotine concentration. Rats trained on this procedure also developed an anxiogenic phenotype. Finally, we assessed alterations in neuroimmune-related gene expression in the medial prefrontal cortex - prelimbic, (mPFC-PL), nucleus accumbens core (AcbC), and ventral tegmental area (VTA). In the AcbC, where α7 expression was increased and β2 was decreased, markers of pro-inflammatory activity were decreased, despite increases in TLR gene expression suggesting that co-use with nicotine modulates inflammatory state downstream from the receptor level. By contrast, in mPFC-PL where α7 was not increased, both TLRs and downstream proinflammatory markers were increased. Taken together, these findings support that there are brain regional and sex differences with co-use of alcohol + nicotine SA and suggest that targeting nicotinic α7 may represent a novel strategy for treating alcohol + nicotine co-dependence.
Collapse
Affiliation(s)
- Christie A Randall
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA
| | - Dongxiao Sun
- Department of Pharmacology, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA
| | - Patrick A Randall
- Department of Anesthesiology and Perioperative Medicine, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA; Department of Pharmacology, Pennsylvania State University, College of Medicine, Hershey PA, 17033, USA.
| |
Collapse
|
3
|
Farrokhi AM, Moshrefi F, Eskandari K, Azizbeigi R, Haghparast A. Hippocampal D1-like dopamine receptor as a novel target for the effect of cannabidiol on extinction and reinstatement of methamphetamine-induced CPP. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111025. [PMID: 38729234 DOI: 10.1016/j.pnpbp.2024.111025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Methamphetamine (METH) is a major health problem without effective pharmacological treatment. Cannabidiol (CBD), a component of the Cannabis sativa plant, is believed to have the potential to inhibit drug-related behavior. However, the neurobiological mechanisms responsible for the effects of CBD remain unclear. Several studies have proposed that the suppressing effects of CBD on drug-seeking behaviors could be through the modulation of the dopamine system. The hippocampus (HIP) D1-like dopamine receptor (D1R) is essential for forming and retrieving drug-associated memory. Therefore, the present study aimed to investigate the role of D1R in the hippocampal CA1 region on the effects of CBD on the extinction and reinstatement of METH-conditioned place preference (CPP). For this purpose, different groups of rats over a 10-day extinction period were administered different doses of intra-CA1 SCH23390 (0.25, 1, or 4 μg/0.5 μl, Saline) as a D1R antagonist before ICV injection of CBD (10 μg/5 μl, DMSO12%). In addition, a different set of animals received intra-CA1 SCH23390 (0.25, 1, or 4 μg/0.5 μl) before CBD injection (50 μg/5 μl) on the reinstatement day. The results revealed that the highest dose of SCH23390 (4 μg) significantly reduced the accelerating effects of CBD on the extinction of METH-CPP (P < 0.01). Furthermore, SCH23390 (1 and 4 μg) in the reinstatement phase notably reversed the preventive effects of CBD on the reinstatement of drug-seeking behavior (P < 0.05 and P < 0.001, respectively). In conclusion, the current study revealed that CBD made a shorter extinction period and suppressed METH reinstatement in part by interacting with D1-like dopamine receptors in the CA1 area of HIP.
Collapse
Affiliation(s)
- Amir Mohammad Farrokhi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Fazel Moshrefi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Kiarash Eskandari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ronak Azizbeigi
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Söderpalm B, Ericson M. Alcohol and the dopamine system. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:21-73. [PMID: 38555117 DOI: 10.1016/bs.irn.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The mesolimbic dopamine pathway plays a major role in drug reinforcement and is likely involved also in the development of drug addiction. Ethanol, like most addictive drugs, acutely activates the mesolimbic dopamine system and releases dopamine, and ethanol-associated stimuli also appear to trigger dopamine release. In addition, chronic exposure to ethanol reduces the baseline function of the mesolimbic dopamine system. The molecular mechanisms underlying ethanol´s interaction with this system remain, however, to be unveiled. Here research on the actions of ethanol in the mesolimbic dopamine system, focusing on the involvement of cystein-loop ligand-gated ion channels, opiate receptors, gastric peptides and acetaldehyde is briefly reviewed. In summary, a great complexity as regards ethanol´s mechanism(s) of action along the mesolimbic dopamine system has been revealed. Consequently, several new targets and possibilities for pharmacotherapies for alcohol use disorder have emerged.
Collapse
Affiliation(s)
- Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Eshel N, Touponse GC, Wang AR, Osterman AK, Shank AN, Groome AM, Taniguchi L, Cardozo Pinto DF, Tucciarone J, Bentzley BS, Malenka RC. Striatal dopamine integrates cost, benefit, and motivation. Neuron 2024; 112:500-514.e5. [PMID: 38016471 PMCID: PMC10922131 DOI: 10.1016/j.neuron.2023.10.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/06/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023]
Abstract
Striatal dopamine (DA) release has long been linked to reward processing, but it remains controversial whether DA release reflects costs or benefits and how these signals vary with motivation. Here, we measure DA release in the nucleus accumbens (NAc) and dorsolateral striatum (DLS) while independently varying costs and benefits and apply behavioral economic principles to determine a mouse's level of motivation. We reveal that DA release in both structures incorporates both reward magnitude and sunk cost. Surprisingly, motivation was inversely correlated with reward-evoked DA release. Furthermore, optogenetically evoked DA release was also heavily dependent on sunk cost. Our results reconcile previous disparate findings by demonstrating that striatal DA release simultaneously encodes cost, benefit, and motivation but in distinct manners over different timescales. Future work will be necessary to determine whether the reduction in phasic DA release in highly motivated animals is due to changes in tonic DA levels.
Collapse
Affiliation(s)
- Neir Eshel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Gavin C Touponse
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Allan R Wang
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Amber K Osterman
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Amei N Shank
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandra M Groome
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lara Taniguchi
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel F Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason Tucciarone
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Brandon S Bentzley
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Ike KGO, Lamers SJC, Kaim S, de Boer SF, Buwalda B, Billeter JC, Kas MJH. The human neuropsychiatric risk gene Drd2 is necessary for social functioning across evolutionary distant species. Mol Psychiatry 2024; 29:518-528. [PMID: 38114631 PMCID: PMC11116113 DOI: 10.1038/s41380-023-02345-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
The Drd2 gene, encoding the dopamine D2 receptor (D2R), was recently indicated as a potential target in the etiology of lowered sociability (i.e., social withdrawal), a symptom of several neuropsychiatric disorders such as Schizophrenia and Major Depression. Many animal species show social withdrawal in response to stimuli, including the vinegar fly Drosophila melanogaster and mice, which also share most human disease-related genes. Here we will test for causality between Drd2 and sociability and for its evolutionary conserved function in these two distant species, as well as assess its mechanism as a potential therapeutic target. During behavioral observations in groups of freely interacting D. melanogaster, Drd2 homologue mutant showed decreased social interactions and locomotor activity. After confirming Drd2's social effects in flies, conditional transgenic mice lacking Drd2 in dopaminergic cells (autoreceptor KO) or in serotonergic cells (heteroreceptor KO) were studied in semi-natural environments, where they could freely interact. Autoreceptor KOs showed increased sociability, but reduced activity, while no overall effect of Drd2 deletion was observed in heteroreceptor KOs. To determine acute effects of D2R signaling on sociability, we also showed that a direct intervention with the D2R agonist Sumanirole decreased sociability in wild type mice, while the antagonist showed no effects. Using a computational ethological approach, this study demonstrates that Drd2 regulates sociability across evolutionary distant species, and that activation of the mammalian D2R autoreceptor, in particular, is necessary for social functioning.
Collapse
Affiliation(s)
- Kevin G O Ike
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sanne J C Lamers
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Soumya Kaim
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sietse F de Boer
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Bauke Buwalda
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jean-Christophe Billeter
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
7
|
Lopez-Cruz L, Phillips BU, Hailwood JM, Saksida LM, Heath CJ, Bussey TJ. Refining the study of decision-making in animals: differential effects of d-amphetamine and haloperidol in a novel touchscreen-automated Rearing-Effort Discounting (RED) task and the Fixed-Ratio Effort Discounting (FRED) task. Neuropsychopharmacology 2024; 49:422-432. [PMID: 37644210 PMCID: PMC10724152 DOI: 10.1038/s41386-023-01707-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
Effort-based decision-making is impaired in multiple psychopathologies leading to significant impacts on the daily life of patients. Preclinical studies of this important transdiagnostic symptom in rodents are hampered, however, by limitations present in currently available decision-making tests, including the presence of delayed reinforcement and off-target cognitive demands. Such possible confounding factors can complicate the interpretation of results in terms of decision-making per se. In this study we addressed this problem using a novel touchscreen Rearing-Effort Discounting (RED) task in which mice choose between two single-touch responses: rearing up to touch an increasingly higher positioned stimulus to obtain a High Reward (HR) or touching a lower stimulus to obtain a Low Reward (LR). To explore the putative advantages of this new approach, RED was compared with a touchscreen version of the well-studied Fixed Ratio-based Effort Discounting (FRED) task, in which multiple touches are required to obtain an HR, and a single response is required to obtain an LR. Results from dopaminergic (haloperidol and d-amphetamine), behavioral (changes in the order of effort demand; fixed-ratio schedule in FRED or response height in RED), and dietary manipulations (reward devaluation by pre-feeding) were consistent with the presence of variables that may complicate interpretation of conventional decision-making tasks, and demonstrate how RED appears to minimize such variables.
Collapse
Affiliation(s)
- Laura Lopez-Cruz
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB, UK.
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK.
| | - Benjamin U Phillips
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB, UK
| | - Jonathan M Hailwood
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB, UK
| | - Lisa M Saksida
- Robarts Research Institute and Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Christopher J Heath
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Timothy J Bussey
- Robarts Research Institute and Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada
| |
Collapse
|
8
|
Havranek T, Bacova Z, Bakos J. Oxytocin, GABA, and dopamine interplay in autism. Endocr Regul 2024; 58:105-114. [PMID: 38656256 DOI: 10.2478/enr-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Oxytocin plays an important role in brain development and is associated with various neurotransmitter systems in the brain. Abnormalities in the production, secretion, and distribution of oxytocin in the brain, at least during some stages of the development, are critical for the pathogenesis of neuropsychiatric diseases, particularly in the autism spectrum disorder. The etiology of autism includes changes in local sensory and dopaminergic areas of the brain, which are also supplied by the hypothalamic sources of oxytocin. It is very important to understand their mutual relationship. In this review, the relationship of oxytocin with several components of the dopaminergic system, gamma-aminobutyric acid (GABA) inhibitory neurotransmission and their alterations in the autism spectrum disorder is discussed. Special attention has been paid to the results describing a reduced expression of inhibitory GABAergic markers in the brain in the context of dopaminergic areas in various models of autism. It is presumed that the altered GABAergic neurotransmission, due to the absence or dysfunction of oxytocin at certain developmental stages, disinhibits the dopaminergic signaling and contributes to the autism symptoms.
Collapse
Affiliation(s)
- Tomas Havranek
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
9
|
Du Y, Zhou S, Ma C, Chen H, Du A, Deng G, Liu Y, Tose AJ, Sun L, Liu Y, Wu H, Lou H, Yu YQ, Zhao T, Lammel S, Duan S, Yang H. Dopamine release and negative valence gated by inhibitory neurons in the laterodorsal tegmental nucleus. Neuron 2023; 111:3102-3118.e7. [PMID: 37499661 DOI: 10.1016/j.neuron.2023.06.021] [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: 11/18/2022] [Revised: 03/25/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023]
Abstract
GABAergic neurons in the laterodorsal tegmental nucleus (LDTGABA) encode aversion by directly inhibiting mesolimbic dopamine (DA). Yet, the detailed cellular and circuit mechanisms by which these cells relay unpleasant stimuli to DA neurons and regulate behavioral output remain largely unclear. Here, we show that LDTGABA neurons bidirectionally respond to rewarding and aversive stimuli in mice. Activation of LDTGABA neurons promotes aversion and reduces DA release in the lateral nucleus accumbens. Furthermore, we identified two molecularly distinct LDTGABA cell populations. Somatostatin-expressing (Sst+) LDTGABA neurons indirectly regulate the mesolimbic DA system by disinhibiting excitatory hypothalamic neurons. In contrast, Reelin-expressing LDTGABA neurons directly inhibit downstream DA neurons. The identification of separate GABAergic subpopulations in a single brainstem nucleus that relay unpleasant stimuli to the mesolimbic DA system through direct and indirect projections is critical for establishing a circuit-level understanding of how negative valence is encoded in the mammalian brain.
Collapse
Affiliation(s)
- Yonglan Du
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Siyao Zhou
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Chenyan Ma
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Hui Chen
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Ana Du
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Guochuang Deng
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yige Liu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; College of Forensic Science, School of Medicine, Xi'an Jiaotong University, No.76, Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Amanda J Tose
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Li Sun
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yijun Liu
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Hangjun Wu
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou 310058, China
| | - Huifang Lou
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yan-Qin Yu
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Ting Zhao
- PKU-Nanjing Joint Institute of Translational Medicine, Nanjing 211800, China
| | - Stephan Lammel
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Shumin Duan
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Hongbin Yang
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
10
|
Passeri A, Municchi D, Cavalieri G, Babicola L, Ventura R, Di Segni M. Linking drug and food addiction: an overview of the shared neural circuits and behavioral phenotype. Front Behav Neurosci 2023; 17:1240748. [PMID: 37767338 PMCID: PMC10520727 DOI: 10.3389/fnbeh.2023.1240748] [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: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Despite a lack of agreement on its definition and inclusion as a specific diagnosable disturbance, the food addiction construct is supported by several neurobiological and behavioral clinical and preclinical findings. Recognizing food addiction is critical to understanding how and why it manifests. In this overview, we focused on those as follows: 1. the hyperpalatable food effects in food addiction development; 2. specific brain regions involved in both food and drug addiction; and 3. animal models highlighting commonalities between substance use disorders and food addiction. Although results collected through animal studies emerged from protocols differing in several ways, they clearly highlight commonalities in behavioral manifestations and neurobiological alterations between substance use disorders and food addiction characteristics. To develop improved food addiction models, this heterogeneity should be acknowledged and embraced so that research can systematically investigate the role of specific variables in the development of the different behavioral features of addiction-like behavior in preclinical models.
Collapse
Affiliation(s)
- Alice Passeri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Diana Municchi
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Giulia Cavalieri
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | | | - Rossella Ventura
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| |
Collapse
|
11
|
Rodríguez-Manzo G, Canseco-Alba A. The endogenous cannabinoid system modulates male sexual behavior expression. Front Behav Neurosci 2023; 17:1198077. [PMID: 37324524 PMCID: PMC10264596 DOI: 10.3389/fnbeh.2023.1198077] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
The endocannabinoid system (ECS) plays a key neuromodulatory role in the brain. Main features of endocannabinoids (eCBs) are that they are produced on demand, in response to enhanced neuronal activity, act as retrograde messengers, and participate in the induction of brain plasticity processes. Sexual activity is a motivated behavior and therefore, the mesolimbic dopaminergic system (MSL) plays a central role in the control of its appetitive component (drive to engage in copulation). In turn, copulation activates mesolimbic dopamine neurons and repeated copulation produces the continuous activation of the MSL system. Sustained sexual activity leads to the achievement of sexual satiety, which main outcome is the transient transformation of sexually active male rats into sexually inhibited animals. Thus, 24 h after copulation to satiety, the sexually satiated males exhibit a decreased sexual motivation and do not respond to the presence of a sexually receptive female with sexual activity. Interestingly, blockade of cannabinoid receptor 1 (CB1R) during the copulation to satiety process, interferes with both the appearance of the long-lasting sexual inhibition and the decrease in sexual motivation in the sexually satiated males. This effect is reproduced when blocking CB1R at the ventral tegmental area evidencing the involvement of MSL eCBs in the induction of this sexual inhibitory state. Here we review the available evidence regarding the effects of cannabinoids, including exogenously administered eCBs, on male rodent sexual behavior of both sexually competent animals and rat sub populations spontaneously showing copulatory deficits, considered useful to model some human male sexual dysfunctions. We also include the effects of cannabis preparations on human male sexual activity. Finally, we review the role played by the ECS in the control of male sexual behavior expression with the aid of the sexual satiety phenomenon. Sexual satiety appears as a suitable model for the study of the relationship between eCB signaling, MSL synaptic plasticity and the modulation of male sexual motivation under physiological conditions that might be useful for the understanding of MSL functioning, eCB-mediated plasticity and their relationship with motivational processes.
Collapse
Affiliation(s)
- Gabriela Rodríguez-Manzo
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav-Sede Sur), Ciudad de México, Mexico
| | - Ana Canseco-Alba
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| |
Collapse
|
12
|
The nucleus accumbens dopamine increase, typically triggered by sexual stimuli in male rats, is no longer produced when animals are sexually inhibited due to sexual satiety. Psychopharmacology (Berl) 2022; 239:3679-3695. [PMID: 36192550 DOI: 10.1007/s00213-022-06240-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/17/2022] [Indexed: 10/10/2022]
Abstract
RATIONALE Exposure of male rats to an inaccessible receptive female and copulation increases dopamine (DA) levels in the nucleus accumbens (NAcc). Males copulating to satiety become sexually inhibited and most of them do not display sexual activity when presented with a sexually receptive female 24 h later. This inhibitory state can be pharmacologically reversed. There are no studies exploring NAcc DA levels during this sexual inhibitory state. OBJECTIVES To characterize changes in NAcc DA and its metabolites' levels during sexual satiety development, during the well-established sexual inhibitory state 24 h later, and during its pharmacological reversal. METHODS Changes in NAcc DA and its metabolites were measured in sexually experienced male rats, using in vivo microdialysis, during copulation to satiety, when presented to a new sexually receptive female 24 h later, and during the pharmacological reversal of the sexual inhibition by anandamide. RESULTS NAcc DA levels remained increased during copulation to satiety. DA basal levels were significantly reduced 24 h after copulation to satiety, as compared to the initial basal levels. Presenting a receptive female behind a barrier 24 h after satiety did not induce the typical NAcc DA elevation in the sexually satiated males but there was a decrease that persisted when they got access to the female, with which they did not copulate. Anandamide injection slightly increased NAcc DA levels coinciding with sexual satiety reversal. CONCLUSIONS Reduced NAcc DA concentrations coincide with the inhibition of an instinctive, natural rewarding behavior suggesting that there might be a DA concentration threshold needed to be responsive to a rewarding stimulus.
Collapse
|
13
|
Canonica T, Zalachoras I. Motivational disturbances in rodent models of neuropsychiatric disorders. Front Behav Neurosci 2022; 16:940672. [PMID: 36051635 PMCID: PMC9426724 DOI: 10.3389/fnbeh.2022.940672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Motivated behavior is integral to the survival of individuals, continuously directing actions toward rewards or away from punishments. The orchestration of motivated behavior depends on interactions among different brain circuits, primarily within the dopaminergic system, that subserve the analysis of factors such as the effort necessary for obtaining the reward and the desirability of the reward. Impairments in motivated behavior accompany a wide range of neuropsychiatric disorders, decreasing the patients’ quality of life. Despite its importance, motivation is often overlooked as a parameter in neuropsychiatric disorders. Here, we review motivational impairments in rodent models of schizophrenia, depression, and Parkinson’s disease, focusing on studies investigating effort-related behavior in operant conditioning tasks and on pharmacological interventions targeting the dopaminergic system. Similar motivational disturbances accompany these conditions, suggesting that treatments aimed at ameliorating motivation levels may be beneficial for various neuropsychiatric disorders.
Collapse
|
14
|
Geurts DEM, den Ouden HEM, Janssen L, Swart JC, Froböse MI, Cools R, Speckens AEM. Aversive Pavlovian inhibition in adult attention-deficit/hyperactivity disorder and its restoration by mindfulness-based cognitive therapy. Front Behav Neurosci 2022; 16:938082. [PMID: 35957921 PMCID: PMC9359138 DOI: 10.3389/fnbeh.2022.938082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Control over the tendency to make or withhold responses guided by contextual Pavlovian information plays a key role in understanding impulsivity and hyperactivity. Here we set out to assess (1) the understudied relation between contextual Pavlovian inhibitory control and hyperactivity/impulsivity in adults with ADHD and (2) whether this inhibition can be enhanced by mindfulness based cognitive therapy (MBCT). Methods Within the framework of a randomized controlled trial 50 Adult ADHD patients were assessed before and after 8 weeks of treatment as usual (TAU) with (n = 24) or without (n = 26) MBCT. We employed a well-established behavioral Pavlovian-to-instrumental transfer task that quantifies Pavlovian inhibitory control over instrumental behavior. Results Task results revealed (1) less aversive Pavlovian inhibition in ADHD patients with clinically relevant hyperactivity/impulsivity than in those without; and (2) enhanced Pavlovian inhibition across all ADHD patients after TAU+MBCT compared with TAU. Conclusion These findings offer new insights in the neurocognitive mechanisms of hyperactivity/impulsivity in ADHD and its treatment: We reveal a role for Pavlovian inhibitory mechanisms in understanding hyperactive/impulsive behaviors in ADHD and point toward MBCT as an intervention that might influence these mechanisms.
Collapse
Affiliation(s)
- Dirk E. M. Geurts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, Netherlands
- *Correspondence: Dirk E. M. Geurts,
| | - Hanneke E. M. den Ouden
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Lotte Janssen
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Jennifer C. Swart
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Monja I. Froböse
- Institute of Experimental Psychology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Anne E. M. Speckens
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
15
|
Chen Y, Zhang L, Ding Z, Wu X, Wang G, Shi J. Effects of 3-methylmethcathinone on conditioned place preference and anxiety-like behavior: Comparison with methamphetamine. Front Mol Neurosci 2022; 15:975820. [PMID: 35935336 PMCID: PMC9354685 DOI: 10.3389/fnmol.2022.975820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
3-Methylmethcathinone (3-MMC), a drug belonging to synthetic cathinones family, raised public attention due to its harmful health effects and abuse potential. Although it has similar properties to other cathinone derivatives, the behavioral effects of 3-MMC remain largely unknown. In the present research, we evaluated the rewarding effect of 3-MMC using conditioned place preference (CPP) paradigm and its effect on anxiety-like behavior using elevated plus maze (EPM) and compared with methamphetamine (METH). Then, we performed a whole-brain c-Fos mapping to identify the specific brain regions in response to 3-MMC exposure and explored the changes of synaptic transmission in nucleus accumbens (NAc) using patch-clamp recording after chronic 3-MMC and METH exposure. 3-MMC induced CPP at higher doses of 3 or 10 mg/kg in rats and acute exposure of 3 mg/kg 3-MMC to rats produced anxiolytic-like effect, while anxiety-like behavior was increased after 7 days of injection with 3-MMC. Whole-brain immunostaining revealed increased c-Fos expression in anterior cingulate cortex (ACC), NAc and ventral tegmental area (VTA) after chronic 3-MMC injection compared with saline, which was similar to METH. Especially, 3-MMC induced more neural activation of VTA compared with METH. Finally, we found that amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) in NAc was decreased after chronic 3-MMC injection, while frequency of sIPSCs and spontaneous excitatory postsynaptic currents (sEPSCs) were not affected. Taken together, our results revealed the addictive potential of 3-MMC and its effect on anxiety-like behavior, which warn the risks of 3-MMC abuse and justify the control of synthetic cathinones. And 3-MMC selectively inhibit inhibitory but not excitatory transmission onto neurons in NAc, which may contribute to its effects.
Collapse
Affiliation(s)
- Yang Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Libo Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
- Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zengbo Ding
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing, China
| | - Guibin Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibin Wang,
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, China
- Jie Shi,
| |
Collapse
|
16
|
A Novel and Selective Dopamine Transporter Inhibitor, (S)-MK-26, Promotes Hippocampal Synaptic Plasticity and Restores Effort-Related Motivational Dysfunctions. Biomolecules 2022; 12:biom12070881. [PMID: 35883437 PMCID: PMC9312958 DOI: 10.3390/biom12070881] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
Dopamine (DA), the most abundant human brain catecholaminergic neurotransmitter, modulates key behavioral and neurological processes in young and senescent brains, including motricity, sleep, attention, emotion, learning and memory, and social and reward-seeking behaviors. The DA transporter (DAT) regulates transsynaptic DA levels, influencing all these processes. Compounds targeting DAT (e.g., cocaine and amphetamines) were historically used to shape mood and cognition, but these substances typically lead to severe negative side effects (tolerance, abuse, addiction, and dependence). DA/DAT signaling dysfunctions are associated with neuropsychiatric and progressive brain disorders, including Parkinson’s and Alzheimer diseases, drug addiction and dementia, resulting in devastating personal and familial concerns and high socioeconomic costs worldwide. The development of low-side-effect, new/selective medicaments with reduced abuse-liability and which ameliorate DA/DAT-related dysfunctions is therefore crucial in the fields of medicine and healthcare. Using the rat as experimental animal model, the present work describes the synthesis and pharmacological profile of (S)-MK-26, a new modafinil analogue with markedly improved potency and selectivity for DAT over parent drug. Ex vivo electrophysiology revealed significantly augmented hippocampal long-term synaptic potentiation upon acute, intraperitoneally delivered (S)-MK-26 treatment, whereas in vivo experiments in the hole-board test showed only lesser effects on reference memory performance in aged rats. However, in effort-related FR5/chow and PROG/chow feeding choice experiments, (S)-MK-26 treatment reversed the depression-like behavior induced by the dopamine-depleting drug tetrabenazine (TBZ) and increased the selection of high-effort alternatives. Moreover, in in vivo microdialysis experiments, (S)-MK-26 significantly increased extracellular DA levels in the prefrontal cortex and in nucleus accumbens core and shell. These studies highlight (S)-MK-26 as a potent enhancer of transsynaptic DA and promoter of synaptic plasticity, with predominant beneficial effects on effort-related behaviors, thus proposing therapeutic potentials for (S)-MK-26 in the treatment of low-effort exertion and motivational dysfunctions characteristic of depression and aging-related disorders.
Collapse
|
17
|
Jiang W, Merhar SL, Zeng Z, Zhu Z, Yin W, Zhou Z, Wang L, He L, Vannest J, Lin W. Neural alterations in opioid-exposed infants revealed by edge-centric brain functional networks. Brain Commun 2022; 4:fcac112. [PMID: 35602654 PMCID: PMC9117006 DOI: 10.1093/braincomms/fcac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Prenatal opioid exposure has been linked to adverse effects spanning multiple neurodevelopmental domains, including cognition, motor development, attention, and vision. However, the neural basis of these abnormalities is largely unknown. A total of 49 infants, including 21 opioid-exposed and 28 controls, were enrolled and underwent MRI (43 ± 6 days old) after birth, including resting state functional MRI. Edge-centric functional networks based on dynamic functional connections were constructed, and machine-learning methods were employed to identify neural features distinguishing opioid-exposed infants from unexposed controls. An accuracy of 73.6% (sensitivity 76.25% and specificity 69.33%) was achieved using 10 times 10-fold cross-validation, which substantially outperformed those obtained using conventional static functional connections (accuracy 56.9%). More importantly, we identified that prenatal opioid exposure preferentially affects inter- rather than intra-network dynamic functional connections, particularly with the visual, subcortical, and default mode networks. Consistent results at the brain regional and connection levels were also observed, where the brain regions and connections associated with visual and higher order cognitive functions played pivotal roles in distinguishing opioid-exposed infants from controls. Our findings support the clinical phenotype of infants exposed to opioids in utero and may potentially explain the higher rates of visual and emotional problems observed in this population. Finally, our findings suggested that edge-centric networks could better capture the neural differences between opioid-exposed infants and controls by abstracting the intrinsic co-fluctuation along edges, which may provide a promising tool for future studies focusing on investigating the effects of prenatal opioid exposure on neurodevelopment.
Collapse
Affiliation(s)
- Weixiong Jiang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Stephanie L. Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital and University of Cincinnati Department of Pediatrics, Cincinnati OH, United States
| | - Zhuohao Zeng
- East Chapel Hill High School, Chapel Hill, North Carolina, United States
| | - Ziliang Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Weiyan Yin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Zhen Zhou
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Li Wang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Lili He
- Department of Radiology, Cincinnati Children’s Hospital and University of Cincinnati, Cincinnati OH, United States
| | - Jennifer Vannest
- Department of Communication Sciences and Disorders, University of Cincinnati, Cincinnati OH, United States
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
18
|
Deane AR, Ward RD. The instrumental role of operant paradigms in translational psychiatric research: Insights from a maternal immune activation model of schizophrenia risk. J Exp Anal Behav 2022; 117:560-575. [PMID: 35319781 PMCID: PMC9314699 DOI: 10.1002/jeab.753] [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: 09/29/2021] [Revised: 02/01/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022]
Abstract
Rigorous behavioral analysis is essential to the translation of research conducted using animal models of neuropsychiatric disease. Here we discuss the use of operant paradigms within our lab as a powerful approach for exploring the biobehavioral bases of disease in the maternal immune activation rat model of schizophrenia. We have investigated a range of disease features in schizophrenia including abnormal perception of time, cognition, learning, motivation, and internal state (psychosis), providing complex insights into brain and behavior. Beyond simple phenotyping, implementing sophisticated operant procedures has been effective in delineating aspects of pathological behavior, identifying interacting pathologies, and isolating contributing mechanisms of disease. We provide comment on the strengths of operant techniques to support high-quality behavioral investigations in fundamental neuropsychiatric research.
Collapse
Affiliation(s)
- Ashley R. Deane
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
| | - Ryan D. Ward
- Department of PsychologyUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
19
|
Moghaddam B, Abbas AI. Depression and Prefrontal Cortex: All Roads Lead to Dopamine. Biol Psychiatry 2022; 91:773-774. [PMID: 35422235 DOI: 10.1016/j.biopsych.2022.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Bita Moghaddam
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon; Department of Psychiatry, Oregon Health and Science University, Portland, Oregon.
| | - Atheir I Abbas
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon; Department of Psychiatry, Oregon Health and Science University, Portland, Oregon; VA Portland Health Care System, Portland, Oregon
| |
Collapse
|
20
|
de la Puente B, Zamanillo D, Romero L, Carceller A, Vela JM, Merlos M, Portillo-Salido E. Comprehensive Preclinical Assessment of Sensory, Functional, Motivational-Affective, and Neurochemical Outcomes in Neuropathic Pain: The Case of the Sigma-1 Receptor. ACS Pharmacol Transl Sci 2022; 5:240-254. [PMID: 35434530 PMCID: PMC9003638 DOI: 10.1021/acsptsci.2c00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Indexed: 12/19/2022]
Abstract
Chronic pain remains a major health problem and is currently facing slow drug innovation. New drug treatments should address not only the sensory-discriminative but also functional and motivational-affective components of chronic pain. In a mouse model of neuropathic pain induced by partial sciatic nerve ligation (PSNL), we analyzed sensory and functional-like outcomes by hindpaw mechanical stimulation and automated gait analysis (CatWalk). We characterized over time a reward-seeking task based on diminished motivation for natural reinforcers (anhedonic-like behavior). To differentiate the appetitive ("wanting") and consummatory ("liking") aspects of motivational behavior, we quantified the latency and number of approaches to eat white chocolate, as well as the eating duration and amount consumed. We explored a putative chronic pain-induced dysregulation of monoamine function by measuring monoamine levels in the nucleus accumbens (NAc), a well-known brain reward area. Finally, we investigated the role of sigma-1 receptor (σ1R) modulation, a nonopioid target, in these multiple dimensions by genetic deletion and pharmacological dose-response studies. After 6 weeks, PSNL increased the approach latency and reduced the consumption of white chocolate in 20-25% of the mice, while around 50-60% had one or the other parameter affected independently. After 10 weeks, sham-operated mice also displayed anhedonic-like behavior. PSNL was associated with reduced extracellular baseline dopamine and increased norepinephrine in the NAc and with a suppression of increased dopamine and serotonin efflux in response to the rewarding stimulus. Genetic and pharmacological blockade of σ1R relieved these multiple alterations in nerve-injured mice. We comprehensively describe sensory, functional, and depression-like impairment of key components of motivated behavior associated with nerve injury. We provide a neurochemical substrate for the depressed mesocorticolimbic reward processing in chronic pain, with a potentially increased translational value. Our results also highlight σ1R for the therapeutic intervention of neuropathic pain.
Collapse
Affiliation(s)
| | - Daniel Zamanillo
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Luz Romero
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Alicia Carceller
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Manuel Merlos
- Welab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | | |
Collapse
|
21
|
Zalachoras I, Astori S, Meijer M, Grosse J, Zanoletti O, de Suduiraut IG, Deussing JM, Sandi C. Opposite effects of stress on effortful motivation in high and low anxiety are mediated by CRHR1 in the VTA. SCIENCE ADVANCES 2022; 8:eabj9019. [PMID: 35319997 PMCID: PMC8942367 DOI: 10.1126/sciadv.abj9019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Individuals frequently differ in their behavioral and cognitive responses to stress. However, whether motivation is differently affected by acute stress in different individuals remains to be established. By exploiting natural variation in trait anxiety in outbred Wistar rats, we show that acute stress facilitates effort-related motivation in low anxious animals, while dampening effort in high anxious ones. This model allowed us to address the mechanisms underlying acute stress-induced differences in motivated behavior. We show that CRHR1 expression levels in dopamine neurons of the ventral tegmental area (VTA)-a neuronal type implicated in the regulation of motivation-depend on animals' anxiety, and these differences in CRHR1 expression levels explain the divergent effects of stress on both effortful behavior and the functioning of mesolimbic DA neurons. These findings highlight CRHR1 in VTA DA neurons-whose levels vary with individuals' anxiety-as a switching mechanism determining whether acute stress facilitates or dampens motivation.
Collapse
Affiliation(s)
- Ioannis Zalachoras
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Corresponding author. (C.S.); (I.Z.); (S.A.)
| | - Simone Astori
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Corresponding author. (C.S.); (I.Z.); (S.A.)
| | - Mandy Meijer
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Isabelle Guillot de Suduiraut
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jan M. Deussing
- Max Planck Institute of Psychiatry/Molecular Neurogenetics, Munich, Germany
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Corresponding author. (C.S.); (I.Z.); (S.A.)
| |
Collapse
|
22
|
Maya-Romero AM, Dodd GE, Landin JD, Zaremba HK, Allen OF, Bilbow MA, Hammaker RD, Santerre-Anderson JL. Adolescent high-fructose corn syrup consumption leads to dysfunction in adult affective behaviors and mesolimbic proteins in male Sprague-Dawley rats. Behav Brain Res 2022; 419:113687. [PMID: 34838930 DOI: 10.1016/j.bbr.2021.113687] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Adolescence is a critical period of development, during which the brain undergoes rapid maturation. Problematically, adolescents are the top consumers of high fructose corn syrup (HFCS) sweetened beverages and snacks, which may have neurodevelopmental consequences. While HFCS consumption has been linked to an increased likelihood of obesity and other physical health impairments, the link between HFCS and persistent behavioral changes is not yet fully established. The present study aimed to assess whether adolescent HFCS consumption could lead to alterations in adult behaviors and protein expression, following cessation. Adolescent HFCS-exposure contributed to deficits in learning and motivation on an effort-related T-Maze procedure, as well as increased immobility time in the forced swim paradigm during adulthood. Molecularly, protracted decreases in accumbal dopamine D1 and D2 receptors and protein kinase G (PKG), as well as increases in tyrosine hydroxylase and GluA2 receptor subunits, were observed following HFCS-exposure. Taken together, these data suggest that adolescent HFCS-consumption leads to protracted dysfunction in affective behaviors and alterations in accumbal proteins which persist following cessation of HFCS-consumption.
Collapse
Affiliation(s)
- Alex M Maya-Romero
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Gina E Dodd
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Justine D Landin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Helen K Zaremba
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Omar F Allen
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Mackenzie A Bilbow
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Rhyce D Hammaker
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA
| | - Jessica L Santerre-Anderson
- Department of Psychology, King's College, Wilkes-Barre, PA, USA; Program in Neuroscience, King's College, Wilkes-Barre, PA, USA.
| |
Collapse
|
23
|
Gamma camera imaging in psychiatric disorders. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00222-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
24
|
Tadayonnejad R, Majid DA, Tsolaki E, Rane R, Wang H, Moody TD, Pauli WM, Pouratian N, Bari AA, Murray SB, O'Doherty JP, Feusner JD. Mesolimbic Neurobehavioral Mechanisms of Reward Motivation in Anorexia Nervosa: A Multimodal Imaging Study. Front Psychiatry 2022; 13:806327. [PMID: 35321230 PMCID: PMC8934777 DOI: 10.3389/fpsyt.2022.806327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/10/2022] [Indexed: 01/04/2023] Open
Abstract
Diminished motivation to pursue and obtain primary and secondary rewards has been demonstrated in anorexia nervosa (AN). However, the neurobehavioral mechanisms underlying the behavioral activation component of aberrant reward motivation remains incompletely understood. This work aims to explore this underexplored facet of reward motivation in AN. We recruited female adolescents with AN, restricting type (n = 32) and a healthy control group (n = 28). All participants underwent functional magnetic resonance imaging (fMRI) while performing a monetary reward task. Diffusion MRI data was also collected to examine the reward motivation circuit's structural connectivity. Behavioral results demonstrated slower speed of reward-seeking behavior in those with AN compared with controls. Accompanying this was lower functional connectivity and reduced white matter structural integrity of the connection between the ventral tegmental area/substantia nigra pars compacta and the nucleus accumbens within the mesolimbic circuit. Further, there was evidence of neurobehavioral decoupling in AN between reward-seeking behavior and mesolimbic regional activation and functional connectivity. Aberrant activity of the bed nucleus of the stria terminalis (BNST) and its connectivity with the mesolimbic system was also evident in AN during the reward motivation period. Our findings suggest functional and structural dysconnectivity within a mesolimbic reward circuit, neurofunctional decoupling from reward-seeking behavior, and abnormal BNST function and circuit interaction with the mesolimbic system. These results show behavioral indicators of aberrant reward motivation in AN, particularly in its activational component. This is mediated neuronally by mesolimbic reward circuit functional and structural dysconnectivity as well as neurobehavioral decoupling. Based on these findings, we suggest a novel circuit-based mechanism of impaired reward processing in AN, with the potential for translation to developing more targeted and effective treatments in this difficult-to-treat psychiatric condition.
Collapse
Affiliation(s)
- Reza Tadayonnejad
- Division of Neuromodulation, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Division of Humanities and Social Sciences, California Institute of Technology, Pasadena, CA, United States
| | - Ds-Adnan Majid
- Cognitive Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Evangelia Tsolaki
- Department of Neursurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Riddhi Rane
- Cognitive Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Huan Wang
- Cognitive Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Teena D Moody
- Cognitive Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Wolfgang M Pauli
- Artificial Intelligence Platform, Microsoft, Redmon, WA, United States
| | - Nader Pouratian
- Department of Neursurgery, University of California, Los Angeles, Los Angeles, CA, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ausaf A Bari
- Department of Neursurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stuart B Murray
- Department of Psychiatry and the Behavioral Sciences, University of Southern California, Los Angeles, CA, United States
| | - John P O'Doherty
- Division of Humanities and Social Sciences, California Institute of Technology, Pasadena, CA, United States.,Computation & Neural Systems Program, California Institute of Technology, Pasadena, CA, United States
| | - Jamie D Feusner
- Cognitive Neuroscience, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Favier M, Carcenac C, Savasta M, Carnicella S. Dopamine D3 Receptors: A Potential Target to Treat Motivational Deficits in Parkinson's Disease. Curr Top Behav Neurosci 2022; 60:109-132. [PMID: 35469394 DOI: 10.1007/7854_2022_316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD), which is traditionally viewed as a motor disorder involving the degeneration of dopaminergic (DA) neurons, has recently been identified as a quintessential neuropsychiatric condition. Indeed, a plethora of non-motor symptoms may occur in PD, including apathy. Apathy can be defined as a lack of motivation or a deficit of goal-directed behaviors and results in a pathological decrease of self-initiated voluntary behavior. Apathy in PD appears to fluctuate with the DA state of the patients, suggesting a critical role of DA neurotransmission in the pathophysiology of this neuropsychiatric syndrome. Using a lesion-based approach, we developed a rodent model which exhibits specific alteration in the preparatory component of motivational processes, reminiscent to apathy in PD. We found a selective decrease of DA D3 receptors (D3R) expression in the dorsal striatum of lesioned rats. Next, we showed that inhibition of D3R neurotransmission in non-lesioned animals was sufficient to reproduce the motivational deficit observed in our model. Interestingly, we also found that pharmacologically targeting D3R efficiently reversed the motivational deficit induced by the lesion. Our findings, among other recent data, suggest a critical role of D3R in parkinsonian apathy and highlight this receptor as a promising target for treating motivational deficits.
Collapse
Affiliation(s)
- Mathieu Favier
- Douglas Mental Health University Institute, McGill University, Verdun, QC, Canada
| | - Carole Carcenac
- Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France
| | - Marc Savasta
- Inserm, Délégation régionale Provence-Alpes-Côte d'Azur et Corse, Marseille CEDEX 09, France
| | - Sebastien Carnicella
- Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble, France.
| |
Collapse
|
26
|
Nomura H, Son C, Aotani D, Shimizu Y, Katsuura G, Noguchi M, Kusakabe T, Tanaka T, Miyazawa T, Hosoda K, Nakao K. Impaired leptin responsiveness in the nucleus accumbens of leptin-overexpressing transgenic mice with dysregulated sucrose and lipid preference independent of obesity. Neurosci Res 2021; 177:94-102. [PMID: 34971637 DOI: 10.1016/j.neures.2021.12.007] [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: 02/24/2021] [Revised: 12/06/2021] [Accepted: 12/26/2021] [Indexed: 11/19/2022]
Abstract
While hypothalamic leptin resistance can occur prior to establishment of obesity, clarification is needed as to whether the impaired response to leptin in the reward-related nuclei occurs independently of obesity. To answer this question, we attempted to dissociate the normally coexisting leptin resistance from obesity. We investigated phenotypes of leptin-overexpressing transgenic mice fed for 1 week with 60 % high-fat diet (HFD) (LepTg-HFD1W mice). After 1 week, we observed that LepTg-HFD1W mice weighed as same as wild type (WT) mice fed standard chow diet (CD) for 1 week (WT-CD1W mice). However, compared to WT-CD1W mice, LepTg-HFD1W mice exhibited attenuated leptin-induced anorexia, decreased leptin-induced c-fos immunostaining in nucleus accumbens (NAc), one of important site of reward system, decreased leptin-stimulated pSTAT3 immunostaining in hypothalamus. Furthermore, neither sucrose nor lipid preference was suppressed by leptin in LepTg-HFD1W mice. On the contrary, leptin significantly suppressed both preferences in WT mice fed HFD (WT-HFD1 W mice). These results indicate that leptin responsiveness decreases in NAc independently of obesity. Additionally, in this situation, suppressive effect of leptin on the hedonic feeding results in impaired regulation. Such findings suggest the impaired leptin responsiveness in NAc partially contributes to dysregulated hedonic feeding behavior independently of obesity.
Collapse
Affiliation(s)
- Hidenari Nomura
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Cheol Son
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Japan.
| | - Daisuke Aotani
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiyuki Shimizu
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Human Health and Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Goro Katsuura
- Department of Social and Behavioral Medicine, Division of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Michio Noguchi
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Kusakabe
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohiro Tanaka
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Miyazawa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kiminori Hosoda
- Department of Human Health and Science, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
27
|
Steidel K, Ruppert MC, Palaghia I, Greuel A, Tahmasian M, Maier F, Hammes J, van Eimeren T, Timmermann L, Tittgemeyer M, Drzezga A, Pedrosa D, Eggers C. Dopaminergic pathways and resting-state functional connectivity in Parkinson's disease with freezing of gait. Neuroimage Clin 2021; 32:102899. [PMID: 34911202 PMCID: PMC8645514 DOI: 10.1016/j.nicl.2021.102899] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/15/2022]
Abstract
Freezing of gait is a common phenomenon of advanced Parkinson's disease. Besides locomotor function per se, a role of cognitive deficits has been suggested. Limited evidence of associated dopaminergic deficits points to caudatal denervation. Further, altered functional connectivity within resting-state networks with importance for cognitive functions has been described in freezers. A potential pathophysiological link between both imaging findings has not yet been addressed. The current study sought to investigate the association between dopaminergic pathway dysintegrity and functional dysconnectivity in relation to FOG severity and cognitive performance in a well-characterized PD cohort undergoing high-resolution 6-[18F]fluoro-L-Dopa PET and functional MRI. The freezing of gait questionnaire was applied to categorize patients (n = 59) into freezers and non-freezers. A voxel-wise group comparison of 6-[18F]fluoro-L-Dopa PET scans with focus on striatum was performed between both well-matched and neuropsychologically characterized patient groups. Seed-to-voxel resting-state functional connectivity maps of the resulting dopamine depleted structures and dopaminergic midbrain regions were created and compared between both groups. For a direct between-group comparison of dopaminergic pathway integrity, a molecular connectivity approach was conducted on 6-[18F]fluoro-L-Dopa scans. With respect to striatal regions, freezers showed significant dopaminergic deficits in the left caudate nucleus, which exhibited altered functional connectivity with regions of the visual network. Regarding midbrain structures, the bilateral ventral tegmental area showed altered functional coupling to regions of the default mode network. An explorative examination of the integrity of dopaminergic pathways by molecular connectivity analysis revealed freezing-associated impairments in mesolimbic and mesocortical pathways. This study represents the first characterization of a link between dopaminergic pathway dysintegrity and altered functional connectivity in Parkinson's disease with freezing of gait and hints at a specific involvement of striatocortical and mesocorticolimbic pathways in freezers.
Collapse
Affiliation(s)
- Kenan Steidel
- Department of Neurology, University Hospital of Marburg, Germany.
| | - Marina C Ruppert
- Department of Neurology, University Hospital of Marburg, Germany; Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Germany
| | - Irina Palaghia
- Department of Neurology, University Hospital of Marburg, Germany
| | - Andrea Greuel
- Department of Neurology, University Hospital of Marburg, Germany
| | - Masoud Tahmasian
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
| | - Franziska Maier
- Department of Psychiatry, University Hospital Cologne, Medical Faculty, Cologne, Germany
| | - Jochen Hammes
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Germany
| | - Thilo van Eimeren
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Germany; Department of Neurology, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn- Cologne, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital of Marburg, Germany; Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany; Cluster of Excellence in Cellular Stress and Aging Associated Disease (CECAD), Cologne, Germany
| | - Alexander Drzezga
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, Medical Faculty and University Hospital Cologne, University Hospital Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn- Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Germany
| | - David Pedrosa
- Department of Neurology, University Hospital of Marburg, Germany; Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Germany
| | - Carsten Eggers
- Department of Neurology, University Hospital of Marburg, Germany; Center for Mind, Brain and Behavior - CMBB, Universities Marburg and Gießen, Germany
| |
Collapse
|
28
|
Courtiol E, Menezes EC, Teixeira CM. Serotonergic regulation of the dopaminergic system: Implications for reward-related functions. Neurosci Biobehav Rev 2021; 128:282-293. [PMID: 34139249 PMCID: PMC8335358 DOI: 10.1016/j.neubiorev.2021.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022]
Abstract
Serotonin is a critical neuromodulator involved in development and behavior. Its role in reward is however still debated. Here, we first review classical studies involving electrical stimulation protocols and pharmacological approaches. Contradictory results on the serotonergic' involvement in reward emerge from these studies. These differences might be ascribable to either the diversity of cellular types within the raphe nuclei or/and the specific projection pathways of serotonergic neurons. We continue to review more recent work, using optogenetic approaches to activate serotonergic cells in the Raphe to VTA pathway. From these studies, it appears that activation of this pathway can lead to reinforcement learning mediated through the excitation of dopaminergic neurons by serotonergic neurons co-transmitting glutamate. Finally, given the importance of serotonin during development on adult emotion, the effect of abnormal early-life levels of serotonin on the dopaminergic system will also be discussed. Understanding the interaction between the serotonergic and dopaminergic systems during development and adulthood is critical to gain insight into the specific facets of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Emmanuelle Courtiol
- Lyon Neuroscience Research Center, UMR 5292- INSERM U1028- Université Lyon 1, 69675 Bron Cedex, France
| | - Edenia C Menezes
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, United States
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, United States; Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
29
|
Cunha C, Smiley JF, Chuhma N, Shah R, Bleiwas C, Menezes EC, Seal RP, Edwards RH, Rayport S, Ansorge MS, Castellanos FX, Teixeira CM. Perinatal interference with the serotonergic system affects VTA function in the adult via glutamate co-transmission. Mol Psychiatry 2021; 26:4795-4812. [PMID: 32398719 PMCID: PMC7657958 DOI: 10.1038/s41380-020-0763-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 11/29/2022]
Abstract
Serotonin and dopamine are associated with multiple psychiatric disorders. How they interact during development to affect subsequent behavior remains unknown. Knockout of the serotonin transporter or postnatal blockade with selective serotonin reuptake inhibitors (SSRIs) leads to novelty-induced exploration deficits in adulthood, potentially involving the dopamine system. Here, we show in the mouse that raphe nucleus serotonin neurons activate ventral tegmental area dopamine neurons via glutamate co-transmission and that this co-transmission is reduced in animals exposed postnatally to SSRIs. Blocking serotonin neuron glutamate co-transmission mimics this SSRI-induced hypolocomotion, while optogenetic activation of dopamine neurons reverses this hypolocomotor phenotype. Our data demonstrate that serotonin neurons modulate dopamine neuron activity via glutamate co-transmission and that this pathway is developmentally malleable, with high serotonin levels during early life reducing co-transmission, revealing the basis for the reduced novelty-induced exploration in adulthood due to postnatal SSRI exposure.
Collapse
Affiliation(s)
- Catarina Cunha
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - John F Smiley
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Nao Chuhma
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Relish Shah
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Cynthia Bleiwas
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Edenia C Menezes
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | - Rebecca P Seal
- Department of Neurobiology and Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, University of California, San Francisco School of Medicine, San Francisco, CA, 94143, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Mark S Ansorge
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Francisco X Castellanos
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA.
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
30
|
Preference for vigorous exercise versus sedentary sucrose drinking: an animal model of anergia induced by dopamine receptor antagonism. Behav Pharmacol 2021; 31:553-564. [PMID: 32141919 DOI: 10.1097/fbp.0000000000000556] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Motivation has activational and directional components. Mesolimbic dopamine is critical for the regulation of behavioral activation and effort-related processes in motivated behaviors. Impairing mesolimbic dopamine function leads to fatigue and anergia, but leaves intact other aspects of reinforce seeking behaviors, such as the consummatory or hedonic component. In male Swiss mice, we characterized the impact of dopamine antagonism on the selection of concurrently presented stimuli that have different vigor requirements. We analyzed running wheel activity versus sucrose solution intake, typically used as a measure of anhedonia. Results are compared with data from nonconcurrent presentation to those stimuli. In the concurrent presentation experiment, control mice preferred to spend time running compared to sucrose intake. Dopamine antagonism shifted relative reinforcer preference, reducing time spent on the running wheel, but actually increasing time-consuming sucrose. Mice increased frequency of bouts for both reinforcers, suggesting that there was fatigue in the running wheel rather than aversion. Moreover, satiation or habituation by preexposing animals to both reinforcers did not shift preferences. In the nonconcurrent experiments, haloperidol reduced running wheel but had no impact on sucrose consumption. Dopamine antagonism did not change preference for sucrose or total volume consumed. Additional correlational analyses indicated that baseline differences in sucrose consumption were independent of baseline running or novelty exploration. Thus, dopamine antagonism seems to have anergic rather than anhedonic effects, and the concurrent presentation in this setting could be useful for assessing preferences based on effort requirements.
Collapse
|
31
|
Fisher ML, Pauly JR, Froeliger B, Turner JR. Translational Research in Nicotine Addiction. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039776. [PMID: 32513669 DOI: 10.1101/cshperspect.a039776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
While commendable strides have been made in reducing smoking initiation and improving smoking cessation rates, current available smoking cessation treatment options are still only mildly efficacious and show substantial interindividual variability in their therapeutic responses. Therefore, the primary goal of preclinical research has been to further the understanding of the neural substrates and genetic influences involved in nicotine's effects and reassess potential drug targets. Pronounced advances have been made by investing in new translational approaches and placing more emphasis on bridging the gap between human and rodent models of dependence. Functional neuroimaging studies have identified key brain structures involved with nicotine-dependence phenotypes such as craving, impulsivity, withdrawal symptoms, and smoking cessation outcomes. Following up with these findings, rodent-modeling techniques have made it possible to dissect the neural circuits involved in these motivated behaviors and ascertain mechanisms underlying nicotine's interactive effects on brain structure and function. Likewise, translational studies investigating single-nucleotide polymorphisms (SNPs) within the cholinergic, dopaminergic, and opioid systems have found high levels of involvement of these neurotransmitter systems in regulating the reinforcing aspects of nicotine in both humans and mouse models. These findings and coordinated efforts between human and rodent studies pave the way for future work determining gene by drug interactions and tailoring treatment options to each individual smoker.
Collapse
Affiliation(s)
- Miranda L Fisher
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| | - James R Pauly
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| | - Brett Froeliger
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky 40536-0596, USA
| |
Collapse
|
32
|
Letzen JE, Boissoneault J, Sevel LS, Robinson ME. Altered mesocorticolimbic functional connectivity in chronic low back pain patients at rest and following sad mood induction. Brain Imaging Behav 2021; 14:1118-1129. [PMID: 30877469 DOI: 10.1007/s11682-019-00076-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Depressive symptoms are common among individuals with chronic pain. Previous work suggests that chronic pain patients have difficulty regulating emotional responses, which is a risk factor for the development of major depressive disorder (MDD). Function of the mesocorticolimbic system, a neural network associated with reward processing, contributes to emotion regulation. This network's dysfunction has been described in chronic pain and MDD research and potentially underlies the relationship among emotion dysregulation, chronic pain, and MDD development. Given that mood induction paradigms have been used to measure emotion regulation, the present study examined intrinsic mesocorticolimbic functional connectivity (FC) after induced sad mood in individuals with and without chronic low back pain (cLBP). Thirty-three MDD-free individuals (17 cLBP) underwent resting-state scanning before and after sad memory-evoked mood induction. A Group [cLBP, healthy control (HC)] x Mood (Neutral, Sadness) repeated measures ANCOVA was conducted on seed-based FC data using a mesolimbic a priori region of interest. Interaction effects were identified in the orbital frontal cortex and inferior frontal gyrus [F(2,29) = 21.07, pFDR < .05. hp2 = .5]. Whereas cLBP showed significantly greater FC between these two regions and the mesolimbic seed under neutral mood, FC among these regions increased in HC and decreased in cLBP under sad mood. Exploratory graph theory analyses further describe between-group differences in mesocorticolimbic network properties. Findings support previous literature describing mesocorticolimbic dysfunction in cLBP and demonstrate aberrant function in emotion regulation. Mesocorticolimbic dysfunction during emotion regulation might contribute to the development of certain depressive phenotypes in chronic pain patients.
Collapse
Affiliation(s)
- Janelle E Letzen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, 5510 Nathan Shock Drive, Suite 100, Baltimore, MD, 21224, USA.
| | - Jeff Boissoneault
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Landrew S Sevel
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| | - Michael E Robinson
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
33
|
Shields AK, Suarez M, Wakabayashi KT, Bass CE. Activation of VTA GABA neurons disrupts reward seeking by altering temporal processing. Behav Brain Res 2021; 410:113292. [PMID: 33836166 DOI: 10.1016/j.bbr.2021.113292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/30/2022]
Abstract
The role of ventral tegmental area (VTA) dopamine in reward, cue processing, and interval timing is well characterized. Using a combinatorial viral approach to target activating DREADDs (Designer Receptors Exclusively Activated by Designer Drugs, hM3D) to GABAergic neurons in the VTA of male rats, we previously showed that activation disrupts responding to reward-predictive cues. Here we explored how VTA GABA neurons influence the perception of time in two fixed interval (FI) tasks, one where the reward or interval is not paired with predictive cues (Non-Cued FI), and another where the start of the FI is signaled by a constant tone that continues until the rewarded response is emitted (Cued FI). Under vehicle conditions in both tasks, responding was characterized by "scalloping" over the 30 s FI, in which responding increased towards the end of the FI. However, when VTA GABA neurons were activated in the Non-Cued FI, the time between the end of the 30 s interval and when the rats made a reinforced response increased. Additionally, post-reinforcement pauses and overall session length increased. In the Cued FI task, VTA GABA activation produced erratic responding, with a decrease in earned rewards. Thus, while both tasks were disrupted by VTA GABA activation, responding that is constrained by a cue was more sensitive to this manipulation, possibly due to convergent effects on timing and cue processing. Together these results demonstrate that VTA GABA activity disrupts the perception of interval timing, particularly when the timing is set by cues.
Collapse
Affiliation(s)
- Andrea K Shields
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, 14214, United States
| | - Mauricio Suarez
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, 14214, United States; Clinical Research Institute on Addictions, University at Buffalo, State University of New York, Buffalo, NY, 14203, United States
| | - Ken T Wakabayashi
- Department of Psychology, University of Nebraska-Lincoln, 1220 T. Street, Lincoln, NE, 68588, United States
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, 14214, United States; Clinical Research Institute on Addictions, University at Buffalo, State University of New York, Buffalo, NY, 14203, United States.
| |
Collapse
|
34
|
Porru S, López-Cruz L, Carratalá-Ros C, Salamone JD, Acquas E, Correa M. Impact of Caffeine on Ethanol-Induced Stimulation and Sensitization: Changes in ERK and DARPP-32 Phosphorylation in Nucleus Accumbens. Alcohol Clin Exp Res 2021; 45:608-619. [PMID: 33471948 DOI: 10.1111/acer.14553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Caffeine is frequently consumed with ethanol to reduce the impairing effects induced by ethanol, including psychomotor slowing or incoordination. Both drugs modulate dopamine (DA)-related markers in accumbens (Acb), and Acb DA is involved in voluntary locomotion and locomotor sensitization. The present study determined whether caffeine can affect locomotion induced by acute and repeated ethanol administration in adult male CD-1 mice. METHODS Acute administration of caffeine (7.5 to 30.0 mg/kg) was evaluated for its effects on acute ethanol-induced (1.5 to 3.5 g/kg) changes in open-field horizontal locomotion, supported rearing, and rearing not supported by the wall. DA receptor-dependent phosphorylation markers were assessed: extracellular signal-regulated kinase (pERK), and dopamine-and cAMP-regulated phosphoprotein Mr32kDa phosphorylated at threonine 75 site (pDARPP-32-Thr75) in Acb core and shell. Acutely administered caffeine was also evaluated in ethanol-sensitized (1.5 g/kg) mice. RESULTS Acute ethanol decreased both types of rearing. Caffeine increased supported rearing but did not block ethanol -induced decreases in rearing. Both substances increased horizontal locomotion in a biphasic manner, and caffeine potentiated ethanol-induced locomotion. Although ethanol administered repeatedly induced sensitization of locomotion and unsupported rearing, acute administration of caffeine to ethanol-sensitized mice in an ethanol-free state resulted in blunted stimulant effects compared with those seen in ethanol-naïve mice. Ethanol increased pERK immunoreactivity in both subregions of the Acb, but coadministration with caffeine blunted this increase. There were no effects on pDARPP-32(Thr75) immunoreactivity. CONCLUSIONS The present results demonstrated that, after the first administration, caffeine potentiated the stimulating actions of ethanol, but did not counteract its suppressant or ataxic effects. Moreover, our results show that caffeine has less activating effects in ethanol-sensitized animals.
Collapse
Affiliation(s)
- Simona Porru
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain.,Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Laura López-Cruz
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| | - Carla Carratalá-Ros
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| | - John D Salamone
- Department of Psychology, University of Connecticut, Storrs, Connecticut, USA
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of Cagliari, Monserrato, Italy
| | - Mercè Correa
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, Castelló, Spain
| |
Collapse
|
35
|
Reduced Motivation in Perinatal Fluoxetine-Treated Mice: A Hypodopaminergic Phenotype. J Neurosci 2021; 41:2723-2732. [PMID: 33536200 DOI: 10.1523/jneurosci.2608-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/17/2022] Open
Abstract
Early life is a sensitive period, in which enhanced neural plasticity allows the developing brain to adapt to its environment. This plasticity can also be a risk factor in which maladaptive development can lead to long-lasting behavioral deficits. Here, we test how early-life exposure to the selective-serotonin-reuptake-inhibitor (SSRI), fluoxetine, affects motivation, and dopaminergic signaling in adulthood. We show for the first time that mice exposed to fluoxetine in the early postnatal period exhibit a reduction in effort-related motivation. These mice also show blunted responses to amphetamine and reduced dopaminergic activation in a sucrose reward task. Interestingly, we find that the reduction in motivation can be rescued in the adult by administering bupropion, a dopamine-norepinephrine reuptake inhibitor used as an antidepressant and a smoke cessation aid but not by fluoxetine. Taken together, our studies highlight the effects of early postnatal exposure of fluoxetine on motivation and demonstrate the involvement of the dopaminergic system in this process.SIGNIFICANCE STATEMENT The developmental period is characterized by enhanced plasticity. During this period, environmental factors have the potential to lead to enduring behavioral changes. Here, we show that exposure to the SSRI fluoxetine during a restricted period in early life leads to a reduction in adult motivation. We further show that this reduction is associated with decreased dopaminergic responsivity. Finally, we show that motivational deficits induced by early-life fluoxetine exposure can be rescued by adult administration of bupropion but not by fluoxetine.
Collapse
|
36
|
Huys QJM, Browning M, Paulus MP, Frank MJ. Advances in the computational understanding of mental illness. Neuropsychopharmacology 2021; 46:3-19. [PMID: 32620005 PMCID: PMC7688938 DOI: 10.1038/s41386-020-0746-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
Computational psychiatry is a rapidly growing field attempting to translate advances in computational neuroscience and machine learning into improved outcomes for patients suffering from mental illness. It encompasses both data-driven and theory-driven efforts. Here, recent advances in theory-driven work are reviewed. We argue that the brain is a computational organ. As such, an understanding of the illnesses arising from it will require a computational framework. The review divides work up into three theoretical approaches that have deep mathematical connections: dynamical systems, Bayesian inference and reinforcement learning. We discuss both general and specific challenges for the field, and suggest ways forward.
Collapse
Affiliation(s)
- Quentin J M Huys
- Division of Psychiatry and Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, UK.
- Camden and Islington NHS Trust, London, UK.
| | - Michael Browning
- Computational Psychiatry Lab, Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Trust, Oxford, UK
| | - Martin P Paulus
- Laureate Institute For Brain Research (LIBR), Tulsa, OK, USA
| | - Michael J Frank
- Cognitive, Linguistic & Psychological Sciences, Neuroscience Graduate Program, Brown University, Providence, RI, USA
- Carney Center for Computational Brain Science, Carney Institute for Brain Science Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| |
Collapse
|
37
|
Moskowitz S, Russ DW, Clark LA, Wages NP, Grooms DR, Woods AJ, Suhr J, Simon JE, O'Shea A, Criss CR, Fadda P, Clark BC. Is impaired dopaminergic function associated with mobility capacity in older adults? GeroScience 2020; 43:1383-1404. [PMID: 33236263 DOI: 10.1007/s11357-020-00303-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/18/2020] [Indexed: 01/14/2023] Open
Abstract
The capacity to move is essential for independence and declines with age. Slow movement speed, in particular, is strongly associated with negative health outcomes. Prior research on mobility (herein defined as movement slowness) and aging has largely focused on musculoskeletal mechanisms and processes. More recent work has provided growing evidence for a significant role of the nervous system in contributing to reduced mobility in older adults. In this article, we report four pieces of complementary evidence from behavioral, genetic, and neuroimaging experiments that, we believe, provide theoretical support for the assertion that the basal ganglia and its dopaminergic function are responsible, in part, for age-related reductions in mobility. We report four a posteriori findings from an existing dataset: (1) slower central activation of ballistic force development is associated with worse mobility among older adults; (2) older adults with the Val/Met intermediate catecholamine-O-methyl-transferase (COMT) genotype involved in dopamine degradation exhibit greater mobility than their homozygous counterparts; (3) there are moderate relationships between performance times from a series of lower and upper extremity tasks supporting the notion that movement speed in older adults is a trait-like attribute; and (4) there is a relationship of functional connectivity within the medial orbofrontal (mOFC) cortico-striatal network and measures of mobility, suggesting that a potential neural mechanism for impaired mobility with aging is the deterioration of the integrity of key regions within the mOFC cortico-striatal network. These findings align with recent basic and clinical science work suggesting that the basal ganglia and its dopaminergic function are mechanistically linked to age-related reductions in mobility capacity.
Collapse
Affiliation(s)
- Simon Moskowitz
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA
| | - David W Russ
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,School of Rehabilitation and Communication Sciences, Ohio University, Athens, OH, USA.,School of Physical Therapy & Rehabilitation Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Leatha A Clark
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,Department of Biomedical Sciences at Ohio University, Athens, OH, USA.,Department of Family Medicine at Ohio University, Athens, OH, USA
| | - Nathan P Wages
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,Department of Biomedical Sciences at Ohio University, Athens, OH, USA
| | - Dustin R Grooms
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,School of Applied Health and Wellness, Ohio University, Athens, OH, USA
| | - Adam J Woods
- Center for Cognitive Aging and Memory, Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Julie Suhr
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,Department of Psychology, Ohio University, Athens, OH, USA
| | - Janet E Simon
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA.,School of Applied Health and Wellness, Ohio University, Athens, OH, USA
| | - Andrew O'Shea
- Center for Cognitive Aging and Memory, Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
| | - Cody R Criss
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA
| | - Paolo Fadda
- Genomics Shared Resource-Comprehensive Cancer Center, The Ohio State University, Athens, OH, USA
| | - Brian C Clark
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, 250 Irvine Hall, Athens, OH, 45701, USA. .,Department of Biomedical Sciences at Ohio University, Athens, OH, USA. .,Division of Geriatric Medicine at Ohio University, Athens, OH, USA.
| |
Collapse
|
38
|
Abstract
Thirst is a highly potent drive that motivates organisms to seek out and consume balance-restoring stimuli. The detection of dehydration is well understood and involves signals of peripheral origin and the sampling of internal milieu by first order homeostatic neurons within the lamina terminalis-particularly glutamatergic neurons of the subfornical organ expressing CaMKIIa (SFOCaMKIIa). However, it remains unknown whether mesolimbic dopamine pathways that are critical for motivation and reinforcement integrate information from these "early" dehydration signals. We used in vivo fiber photometry in the ventral tegmental area and measured phasic dopamine responses to a water-predictive cue. Thirst, but not hunger, potentiated the phasic dopamine response to the water cue. In euvolemic rats, the dipsogenic hormone angiotensin II, but not the orexigenic hormone ghrelin, potentiated the dopamine response similarly to that observed in water-deprived rats. Chemogenetic manipulations of SFOCaMKIIa revealed bidirectional control of phasic dopamine signaling during cued water reward. Taking advantage of within-subject designs, we found predictive relationships between changes in cue-evoked dopamine response and changes in behavioral responses-supporting a role for dopamine in motivation induced by homeostatic need. Collectively, we reveal a putative mechanism for the invigoration of goal-directed behavior: internal milieu communicates to first order, need state-selective circuits to potentiate the mesolimbic dopamine system's response to cues predictive of restorative stimuli.
Collapse
|
39
|
Augustin SM, Loewinger GC, O'Neal TJ, Kravitz AV, Lovinger DM. Dopamine D2 receptor signaling on iMSNs is required for initiation and vigor of learned actions. Neuropsychopharmacology 2020; 45:2087-2097. [PMID: 32811899 PMCID: PMC7547091 DOI: 10.1038/s41386-020-00799-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Striatal dopamine D2 receptors (D2Rs) are important for motor output. Selective deletion of D2Rs from indirect pathway-projecting medium spiny neurons (iMSNs) impairs locomotor activities in a task-specific manner. However, the role of D2Rs in the initiation of motor actions in reward seeking and taking is not fully understood, and there is little information about how receptors contribute under different task demands and with different outcome types. The iMSN-D2Rs modulate neuronal activity and synaptic transmission, exerting control on circuit functions that may play distinct roles in action learning and performance. Selective deletion of D2Rs on iMSNs resulted in slower action initiation and response rate in an instrumental conditioning task, but only when performance demand was increased. The iMSN-Drd2KO mice were also slower to initiate swimming in a T-maze procedural learning task but were unimpaired in cognitive function and behavioral flexibility. In contrast, in a Pavlovian discrimination learning task, iMSN-Drd2KO mice exhibited normal acquisition and extinction of rewarded responding. The iMSN-Drd2KO mice showed performance deficits at all phases of rotarod skill learning. These findings reveal that dopamine modulation through iMSN-D2Rs influences the ability to self-initiate actions, as well as the willingness and/or vigor with which these responses are performed. However, these receptors seem to have little influence on simple associative learning or on stimulus-driven responding. The loss of normal D2R roles may contribute to disorders in which impaired dopamine signaling leads to hypokinesia or impaired initiation of specific voluntary actions.
Collapse
Affiliation(s)
- Shana M Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Gabriel C Loewinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Timothy J O'Neal
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Graduate Program in Neuroscience and Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington, 98195, USA
| | - Alexxai V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
- Departments of Psychiatry, Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA.
| |
Collapse
|
40
|
Liu Y, Montgomery SE, Juarez B, Morel C, Zhang S, Kong Y, Calipari ES, Nestler EJ, Zhang L, Han MH. Different adaptations of dopamine release in Nucleus Accumbens shell and core of individual alcohol drinking groups of mice. Neuropharmacology 2020; 175:108176. [PMID: 32497591 PMCID: PMC7492398 DOI: 10.1016/j.neuropharm.2020.108176] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 01/05/2023]
Abstract
Alcohol use disorder (AUD) places a tremendous burden on society, with approximately two billion alcohol users in the world. While most people drink alcohol recreationally, a subpopulation (3-5%) engages in reckless and compulsive drinking, leading to the development of AUD and alcohol dependence. The Ventral Tegmental Area (VTA)-Nucleus Accumbens (NAc) circuit has been shown to encode rewarding stimuli and drive individual alcohol drinking behavior. Our previous work successfully separated C57BL/6J isogenic mice into high or low alcohol drinking subgroups after a 12-day, two-bottle choice voluntary alcohol access paradigm. Electrophysiological studies revealed that low alcohol drinking mice exhibited elevated spontaneous and burst firing properties of their VTA dopamine (DA) neurons and specifically mimicking this pattern of activity in VTA-NAc neurons in high alcohol drinking mice using optogenetics decreased their alcohol preference. It is also known that VTA DA neurons encode the salience and rewarding properties of external stimuli while also regulating downstream dopamine concentrations. Here, as a follow-up to this study, we utilized Fast Scan Cyclic Voltammetry (FSCV) to examine dopamine release in the NAc shell and core between alcohol drinking groups. We observed dynamic changes of dopamine release in the core of high drinking mice, but failed to see widely significant differences of dopamine release in the shell of both groups, when compared with ethanol-naive controls. Overall, the present data suggest subregion-specific differences of evoked dopamine release in the NAc of low and high alcohol drinking mice, and may provide an anatomical substrate for individual alcohol drinking behavior. This article is part of the special issue on Stress, Addiction and Plasticity.
Collapse
Affiliation(s)
- Yutong Liu
- Key Laboratory of Functional Proteomics of Guangdong Province, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah E Montgomery
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Barbara Juarez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry & Behavioral Sciences, University of Washington, USA
| | - Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Song Zhang
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yimeng Kong
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Erin S Calipari
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt Brain Institute, Vanderbilt University, Nashville TN, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
41
|
Ohmann HA, Kuper N, Wacker J. A low dosage of the dopamine D2-receptor antagonist sulpiride affects effort allocation for reward regardless of trait extraversion. PERSONALITY NEUROSCIENCE 2020; 3:e7. [PMID: 32656492 PMCID: PMC7327436 DOI: 10.1017/pen.2020.7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022]
Abstract
Dopamine (DA) is known to be involved in various aspects of reward processing and goal-directed behavior. The present preregistered study aims at directly accessing the causal influence of DA activity on reward motivation in humans, while also accounting for trait extraversion. Therefore, we examined the effect of a single dose of the DA D2 receptor antagonist sulpiride (200 mg) on effort allocation in a modified version of the Effort-Expenditure for Reward Task (EEfRT). Based on its presumably DA increasing action, we expected the low dose of sulpiride to increase participants' willingness to allocate effort during the modified EEfRT relative to placebo, especially in trials with low probability of reward attainment. Further, we expected a moderating effect of trait extraversion on the effects of sulpiride. Two hundred and three healthy male participants were tested in a randomized, double-blind between-subjects design. Contrary to our expectations, sulpiride reduced the average number of clicks within the modified EEfRT and did not interact with reward attributes, suggesting a more global and not reward-specific effect of sulpiride. Furthermore, trait extraversion did not moderate the effect of sulpiride. Our results provide initial support for the validity of the modified version of the EEfRT, suggesting a possible inhibiting effect of a low dose of sulpiride on approach motivation regardless of trait extraversion. However, given the mixed pattern of findings and the possible confounding role of motoric abilities, further studies examining these effects are clearly warranted.
Collapse
Affiliation(s)
- Hanno Andreas Ohmann
- Faculty of Psychology and Human Movement Science, Universität Hamburg, Hamburg, Germany
| | - Niclas Kuper
- Faculty of Psychology and Human Movement Science, Universität Hamburg, Hamburg, Germany
- Faculty of Psychology and Sports Science, Universität Bielefeld, Bielefeld, Germany
| | - Jan Wacker
- Faculty of Psychology and Human Movement Science, Universität Hamburg, Hamburg, Germany
| |
Collapse
|
42
|
Collo G, Mucci A, Giordano GM, Merlo Pich E, Galderisi S. Negative Symptoms of Schizophrenia and Dopaminergic Transmission: Translational Models and Perspectives Opened by iPSC Techniques. Front Neurosci 2020; 14:632. [PMID: 32625059 PMCID: PMC7315891 DOI: 10.3389/fnins.2020.00632] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/22/2020] [Indexed: 12/18/2022] Open
Abstract
Negative symptoms (NS) represent a heterogeneous dimension of schizophrenia (SCZ), associated with a poor functional outcome. A dysregulated dopamine (DA) system, including a reduced D1 receptor activation in the prefrontal cortex, DA hypoactivity in the caudate and alterations in D3 receptor activity, seems to contribute to the pathogenesis of NS. However, failure to take into account the NS heterogeneity has slowed down progress in research on their neurobiological correlates and discoveries of new effective treatments. A better neurobiological characterization of NS is needed, and this requires objective quantification of their features that can be applied in translational models, such as animal models and human inducible pluripotent stem cells (iPSC). In this review we summarize the evidence for dopaminergic alterations relevant to NS in translational animal models focusing on dysfunctional motivation, a core aspect of NS. Among others, experiments on mutant rodents with an overexpression of DA D2 or D3 receptors and the dopamine deficient mice are discussed. In the second part we summarize the findings from recent studies using iPSC to model the pathogenesis of SCZ. By retaining the genetic background of risk genetic variants, iPSC offer the possibility to study the effect of de novo mutations or inherited polymorphisms from subgroups of patients and their response to drugs, adding an important tool for personalized psychiatry. Given the key role of DA in NS, we focus on findings of iPSC-derived DA neurons. Since implementation of iPSC-derived neurons to study the neurobiology of SCZ is a relatively recent acquisition, the available data are limited. We highlight some methodological aspects of relevance in the interpretation of in vitro testing results, including limitations and strengths, offering a critical viewpoint for the implementation of future pharmacological studies aimed to the discovery and characterization of novel treatments for NS.
Collapse
Affiliation(s)
- Ginetta Collo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Armida Mucci
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giulia M. Giordano
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Emilio Merlo Pich
- Research & Development, Alfasigma Schweiz, Zofingen, Switzerland
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Silvana Galderisi
- Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
43
|
Sagheddu C, Pintori N, Kalaba P, Dragačević V, Piras G, Lubec J, Simola N, De Luca MA, Lubec G, Pistis M. Neurophysiological and Neurochemical Effects of the Putative Cognitive Enhancer ( S)-CE-123 on Mesocorticolimbic Dopamine System. Biomolecules 2020; 10:biom10050779. [PMID: 32443397 PMCID: PMC7277835 DOI: 10.3390/biom10050779] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Treatments for cognitive impairments associated with neuropsychiatric disorders, such as attention deficit hyperactivity disorder or narcolepsy, aim at modulating extracellular dopamine levels in the brain. CE-123 (5-((benzhydrylsulfinyl)methyl) thiazole) is a novel modafinil analog with improved specificity and efficacy for dopamine transporter inhibition that improves cognitive and motivational processes in experimental animals. We studied the neuropharmacological and behavioral effects of the S-enantiomer of CE-123 ((S)-CE-123) and R-modafinil in cognitive- and reward-related brain areas of adult male rats. In vivo single unit recordings in anesthetized animals showed that (S)-CE-123, but not R-modafinil, dose-dependently (1.25 to 10 mg/kg i.v.) reduced firing of pyramidal neurons in the infralimbic/prelimbic (IL/PrL) cortex. Neither compound the affected firing activity of ventral tegmental area dopamine cells. In freely moving animals, (S)-CE-123 (10 mg/kg i.p.) increased extracellular dopamine levels in the IL/PrL, with different patterns when compared to R-modafinil (10 mg/kg i.p.); in the nucleus accumbens shell, a low and transitory increase of dopamine was observed only after (S)-CE-123. Neither (S)-CE-123 nor R-modafinil initiated the emission of 50-kHz ultrasonic vocalizations, a behavioral marker of positive affect and drug-mediated reward. Our data support previous reports of the procognitive effects of (S)-CE-123, and show a minor impact on reward-related dopaminergic areas.
Collapse
Affiliation(s)
- Claudia Sagheddu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Nicholas Pintori
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Vladimir Dragačević
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1010 Vienna, Austria; (P.K.); (V.D.)
| | - Gessica Piras
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Jana Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Nicola Simola
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| | - Marco Pistis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy; (C.S.); (N.P.); (G.P.); (N.S.); (M.A.D.L.)
- Neuroscience Institute, National Research Council of Italy (CNR), Section of Cagliari, 09100 Cagliari, Italy
- Correspondence: (G.L.); (M.P.); Tel.: +43-(0)-6622420-0 (G.L.); +39-070-675-4324 (M.P.)
| |
Collapse
|
44
|
Kotarska A, Fernandes L, Kleene R, Schachner M. Cell adhesion molecule close homolog of L1 binds to the dopamine receptor D2 and inhibits the internalization of its short isoform. FASEB J 2020; 34:4832-4851. [PMID: 32052901 DOI: 10.1096/fj.201900577rrrr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 02/05/2023]
Abstract
Cell adhesion molecule close homolog of L1 (CHL1) and the dopamine receptor D2 (DRD2) are associated with psychiatric and mental disorders. We here show that DRD2 interacts with CHL1 in mouse brain, as evidenced by co-immunostaining, proximity ligation assay, co-immunoprecipitation, and pull-down assay with recombinant extracellular CHL1 domain fused to Fc (CHL1-Fc). Direct binding of CHL1-Fc to the first extracellular loop of DRD2 was shown by ELISA. Using HEK cells transfected to co-express CHL1 and the short (DRD2-S) or long (DRD2-L) DRD2 isoforms, co-localization of CHL1 and both isoforms was observed by immunostaining and proximity ligation assay. Moreover, CHL1 inhibited agonist-triggered internalization of DRD2-S. Proximity ligation assay showed close interaction between CHL1 and DRD2 in neurons expressing dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP32) or tyrosine hydroxylase (TH) in tissue sections of adult mouse striatum. In cultures of striatum or ventral midbrain, CHL1 was also closely associated with DRD2 in DARPP32- or TH-immunopositive cells, respectively. In the dorsal striatum of CHL1-deficient mice, lower levels of DRD2 and phosphorylated TH were observed, when compared to wild-type littermates. In the ventral striatum of CHL1-deficient mice, levels of phosphorylated DARPP32 were reduced. We propose that CHL1 regulates DRD2-dependent presynaptic and postsynaptic functions.
Collapse
Affiliation(s)
- Agnieszka Kotarska
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
45
|
Li Y, Ruan Y, He Y, Cai Q, Pan X, Zhang Y, Liu C, Pu Z, Yang J, Chen M, Huang L, Zhou J, Chen JF. Striatopallidal adenosine A 2A receptors in the nucleus accumbens confer motivational control of goal-directed behavior. Neuropharmacology 2020; 168:108010. [PMID: 32061899 DOI: 10.1016/j.neuropharm.2020.108010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/25/2022]
Abstract
The ability to learn the reward-value and action-outcome contingencies in dynamic environment is critical for flexible adaptive behavior and development of effective pharmacological control of goal-directed behaviors represents an important challenge for improving the deficits in goal-directed behavior which may underlie seemingly disparate symptoms across psychiatric disorders. Adenosine A2A receptor (A2AR) is emerging as a novel neuromodulatory target for controlling goal-directed behavior for its unique neuromodulatory features: the ability to integrate dopamine and glutamate signaling, the "brake" constraint of various cognitive processes and the balanced control of goal-directed and habit actions. However, the contribution and circuit mechanisms of the striatopallidal A2ARs in nucleus accumbens (NAc) to control of goal-directed behavior remain to be determined. Here, we employed newly developed opto-A2AR and the focal A2AR knockdown strategies to demonstrate the causal role of NAc A2AR in control of goal-directed behavior. Furthermore, we dissected out multiple distinct behavioral mechanisms underlying which NAc A2ARs control goal-directed behavior: (i) NAc A2ARs preferentially control goal-directed behavior at the expense of habit formation. (ii) NAc A2ARs modify the animals' sensitivity to the value of the reward without affecting the action-outcome contingency. (iii) A2AR antagonist KW6002 promotes instrumental actions by invigorating motivation. (iv) NAc A2ARs facilitate Pavlovian incentive value transferring to instrumental action. (v) NAc A2ARs control goal-directed behavior probably not through NAc-VP pathway. These insights into the behavioral and circuit mechanisms for NAc A2AR control of goal-directed behavior facilitate translational potential for A2AR antagonists in reversal of deficits in goal-directed decision-making associated with multiple neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Yang Ruan
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Yan He
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Qionghui Cai
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Xinran Pan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Road, Wenzhou, Zhejiang, 325027, China; The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Yu Zhang
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Chengwei Liu
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Zhilan Pu
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Jingjing Yang
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Mozi Chen
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Linshan Huang
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Jianhong Zhou
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China
| | - Jiang-Fan Chen
- The Institute of Molecular Medicine, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
46
|
Papilloud A, Weger M, Bacq A, Zalachoras I, Hollis F, Larrieu T, Battivelli D, Grosse J, Zanoletti O, Parnaudeau S, Tronche F, Sandi C. The glucocorticoid receptor in the nucleus accumbens plays a crucial role in social rank attainment in rodents. Psychoneuroendocrinology 2020; 112:104538. [PMID: 31841985 DOI: 10.1016/j.psyneuen.2019.104538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/01/2019] [Accepted: 11/29/2019] [Indexed: 01/05/2023]
Abstract
Social hierarchy in social species is usually established through competitive encounters with conspecifics. It determines the access to limited resources and, thus, leads to reduced fights among individuals within a group. Despite the known importance of social rank for health and well-being, the knowledge about the processes underlying rank attainment remains limited. Previous studies have highlighted the nucleus accumbens (NAc) as a key brain region in the attainment of social hierarchies in rodents. In addition, glucocorticoids and the glucocorticoid receptor (GR) have been implicated in the establishment of social hierarchies and social aversion. However, whether GR in the NAc is involved in social dominance is not yet known. To address this question, we first established that expression levels of GR in the NAc of high anxious, submissive-prone rats are lower than that of their low anxious, dominant-prone counterparts. Furthermore, virally-induced downregulation of GR expression in the NAc in rats led to an improvement of social dominance rank. We found a similar result in a cell-specific mouse model lacking GR in dopaminoceptive neurons (i.e., neurons containing dopamine receptors). Indeed, when cohabitating in dyads of mixed genotypes, mice deficient for GR in dopaminoceptive neurons had a higher probability to become dominant than wild-type mice. Overall, our results highlight GR in the NAc and in dopaminoceptive neurons as an important regulator of social rank attainment.
Collapse
Affiliation(s)
- Aurélie Papilloud
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Meltem Weger
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Alexandre Bacq
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Ioannis Zalachoras
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Fiona Hollis
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Thomas Larrieu
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Dorian Battivelli
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Sébastien Parnaudeau
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - François Tronche
- Neuroscience Paris-Seine, Gene Regulation and Adaptive Behaviors Team, CNRS UMR8246 - INSERM U1130 - Sorbonne Université, Institut De Biologie Paris-Seine, Paris, France
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland.
| |
Collapse
|
47
|
Exercise-Induced Adaptations to the Mouse Striatal Adenosine System. Neural Plast 2020; 2020:5859098. [PMID: 32399024 PMCID: PMC7204111 DOI: 10.1155/2020/5859098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Adenosine acts as a key regulator of striatum activity, in part, through the antagonistic modulation of dopamine activity. Exercise can increase adenosine activity in the brain, which may impair dopaminergic functions in the striatum. Therefore, long-term repeated bouts of exercise may subsequently generate plasticity in striatal adenosine systems in a manner that promotes dopaminergic activity. This study investigated the effects of long-term voluntary wheel running on adenosine 1 (A1R), adenosine 2A (A2AR), dopamine 1 (D1R), and dopamine 2 (D2R) receptor protein expression in adult mouse dorsal and ventral striatum structures using immunohistochemistry. In addition, equilibrative nucleoside transporter 1 (ENT1) protein expression was examined after wheel running, as ENT1 regulates the bidirectional flux of adenosine between intra- and extracellular space. The results suggest that eight weeks of running wheel access spared age-related increases of A1R and A2AR protein concentrations across the dorsal and ventral striatal structures. Wheel running mildly reduced ENT1 protein levels in ventral striatum subregions. Moreover, wheel running mildly increased D2R protein density within striatal subregions in the dorsal medial striatum, nucleus accumbens core, and the nucleus accumbens shell. However, D1R protein expression in the striatum was unchanged by wheel running. These data suggest that exercise promotes adaptations to striatal adenosine systems. Exercise-reduced A1R and A2AR and exercise-increased D2R protein levels may contribute to improved dopaminergic signaling in the striatum. These findings may have implications for cognitive and behavioral processes, as well as motor and psychiatric diseases that involve the striatum.
Collapse
|
48
|
Brandão AF, Bonet IJM, Pagliusi M, Zanetti GG, Pho N, Tambeli CH, Parada CA, Vieira AS, Sartori CR. Physical Activity Induces Nucleus Accumbens Genes Expression Changes Preventing Chronic Pain Susceptibility Promoted by High-Fat Diet and Sedentary Behavior in Mice. Front Neurosci 2020; 13:1453. [PMID: 32038148 PMCID: PMC6987254 DOI: 10.3389/fnins.2019.01453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
Recent findings from rodent studies suggest that high-fat diet (HFD) increases hyperalgesia independent of obesity status. Furthermore, weight loss interventions such as voluntary physical activity (PA) for adults with obesity or overweight was reported to promote pain reduction in humans with chronic pain. However, regardless of obesity status, it is not known whether HFD intake and sedentary (SED) behavior is underlies chronic pain susceptibility. Moreover, differential gene expression in the nucleus accumbens (NAc) plays a crucial role in chronic pain susceptibility. Thus, the present study used an adapted model of the inflammatory prostaglandin E2 (PGE2)-induced persistent hyperalgesia short-term (PH-ST) protocol for mice, an HFD, and a voluntary PA paradigm to test these hypotheses. Therefore, we performed an analysis of differential gene expression using a transcriptome approach of the NAc. We also applied a gene ontology enrichment tools to identify biological processes associated with chronic pain susceptibility and to investigate the interaction between the factors studied: diet (standard diet vs. HFD), physical activity behavior (SED vs. PA) and PH-ST (PGE vs. saline). Our results demonstrated that HFD intake and sedentary behavior promoted chronic pain susceptibility, which in turn was prevented by voluntary physical activity, even when the animals were fed an HFD. The transcriptome of the NAc found 2,204 differential expression genes and gene ontology enrichment analysis revealed 41 biologic processes implicated in chronic pain susceptibility. Taking these biological processes together, our results suggest that genes related to metabolic and mitochondria stress were up-regulated in the chronic pain susceptibility group (SED-HFD-PGE), whereas genes related to neuroplasticity were up-regulated in the non-chronic pain susceptibility group (PA-HFD-PGE). These findings provide pieces of evidence that HFD intake and sedentary behavior provoked gene expression changes in the NAc related to promotion of chronic pain susceptibility, whereas voluntary physical activity provoked gene expression changes in the NAc related to prevention of chronic pain susceptibility. Finally, our findings confirmed previous literature supporting the crucial role of voluntary physical activity to prevent chronic pain and suggest that low levels of voluntary physical activity would be helpful and highly recommended as a complementary treatment for those with chronic pain.
Collapse
Affiliation(s)
- Arthur Freitas Brandão
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Ivan José Magayewski Bonet
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Marco Pagliusi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Gabriel Gerardini Zanetti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Nam Pho
- eScience Institute, University of Washington, Seattle, WA, United States
| | - Cláudia Herrera Tambeli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Cesar Renato Sartori
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
49
|
Complementary Control over Habits and Behavioral Vigor by Phasic Activity in the Dorsolateral Striatum. J Neurosci 2020; 40:2139-2153. [PMID: 31969469 DOI: 10.1523/jneurosci.1313-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Despite clear evidence linking the basal ganglia to the control of outcome insensitivity (i.e., habit) and behavioral vigor (i.e., its behavioral speed/fluidity), it remains unclear whether or how these functions relate to one another. Here, using male Long-Evans rats in response-based and cue-based maze-running tasks, we demonstrate that phasic dorsolateral striatum (DLS) activity occurring at the onset of a learned behavior regulates how vigorous and habitual it is. In a response-based task, brief optogenetic excitation at the onset of runs decreased run duration and the occurrence of deliberative behaviors, whereas midrun stimulation carried little effect. Outcome devaluation showed these runs to be habitual. DLS inhibition at run start did not produce robust effects on behavior until after outcome devaluation. At that time, when the DLS was plausibly most critically required for performance (i.e., habitual), inhibition reduced performance vigor measures and caused a dramatic loss of habitual responding (i.e., animals quit the task). In a second cue-based "beacon" task requiring behavior initiation at the start of the run and again in the middle of the run, DLS excitation at both time points could improve the vigor of runs. Postdevaluation testing showed behavior on the beacon task to be habitual as well. This pattern of results suggests that one role for phasic DLS activity at behavior initiation is to promote the execution of the behavior in a vigorous and habitual fashion by a diverse set of measures.SIGNIFICANCE STATEMENT Our research expands the literature twofold. First, we find that features of a habitual behavior that are typically studied separately (i.e., maze response performance, deliberation movements, running vigor, and outcome insensitivity) are quite closely linked together. Second, efforts have been made to understand "what" the dorsolateral striatum (DLS) does for habitual behavior, and our research provides a key set of results showing "when" it is important (i.e., at behavior initiation). By showing such dramatic control over habits by DLS activity in a phasic time window, plausible real-world applications could involve more informed DLS perturbations to curb intractably problematic habits.
Collapse
|
50
|
Correia C, Romieu P, Olmstead MC, Befort K. Can cocaine-induced neuroinflammation explain maladaptive cocaine-associated memories? Neurosci Biobehav Rev 2020; 111:69-83. [PMID: 31935376 DOI: 10.1016/j.neubiorev.2020.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/01/2020] [Indexed: 12/19/2022]
Abstract
Persistent and intrusive memories define a number of psychiatric disorders, including posttraumatic stress disorder and substance use disorder. In the latter, memory for drug-paired cues plays a critical role in sustaining compulsive drug use as these are potent triggers of relapse. As with many drugs, cocaine-cue associated memory is strengthened across presentations as cues become reliable predictors of drug availability. Recently, the targeting of cocaine-associated memory through disruption of the reconsolidation process has emerged as a potential therapeutic strategy; reconsolidation reflects the active process by which memory is re-stabilized after retrieval. In addition, a separate line of work reveals that neuroinflammatory markers, regulated by cocaine intake, play a role in memory processes. Our review brings these two literatures together by summarizing recent findings on cocaine-associated reconsolidation and cocaine-induced neuroinflammation. We discuss the interactions between reconsolidation processes and neuroinflammation following cocaine use, concluding with a new perspective on treatment to decrease risk of relapse to cocaine use.
Collapse
Affiliation(s)
- Caroline Correia
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Pascal Romieu
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Mary C Olmstead
- Dept. Psychology, Centre for Neuroscience Studies, Queen's University, Kingston ON, K7L 3N6, Canada
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France.
| |
Collapse
|